1
|
Bratosiewicz-Wąsik J, Wąsik TJ. Genetic variants of IL-10 promoter influence susceptibility to HIV-1 infection and disease progression in the Polish population: IL-10 polymorphisms and HIV-1. Hum Immunol 2024; 85:111086. [PMID: 39153369 DOI: 10.1016/j.humimm.2024.111086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 08/19/2024]
Abstract
The risk of HIV-1 infection and the rate of disease progression vary considerably among individuals and the genetic makeup of the host may be one of the possible reasons for this. We aimed to determine association of functional single nucleotide polymorphism (SNPs), -1082A/G (rs1800896), -819C/T (rs1800871), and -592C/A (rs1800872) in IL-10 gene, with the susceptibility to HIV-1 infection and clinical parameters expressed as a baseline CD4+ T cell count, CD8+ T cell count, and viral load. Therapy naïve HIV-1 infected individuals and HIV-1 seronegative controls from Poland were recruited for this study. Genotyping results revealed significantly higher frequency of -1082G/G genotype (28.1 % vs 16.1 %; p = 0.0019, OR=0.49) and -1082G allele (47.6 % vs 38.8 %; p = 0.0028, OR = 0.70) as well as lower frequency of -592 and -819 heterozygosity (45.0 % vs 34.4 %; p = 0.0266, OR = 1.47) in controls compared to seropositive subjects. High producing haplotype GCC was associated with increased risk of HIV-1 infection (p = 0.0018, OR = 1.52). Individuals possessing -592 and -819 minor allele had significantly higher CD8+ T cell count compared to the wild type allele carriers (p = 0.0303). Moreover, presence of -1082G allele was related with lower viral load as well as CD4+ and CD8+ T cells counts among patients infected with R5 HIV-1 variant. Thus, IL-10 gene promoter variants may be a risk factor for HIV-1 transmission and may modulate disease progression in the Polish population.
Collapse
Affiliation(s)
- Jolanta Bratosiewicz-Wąsik
- Department of Microbiology and Virology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, ul. Jagiellońska 4, Poland.
| | - Tomasz J Wąsik
- Department of Medical Microbiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, 40-752 Katowice, ul. Medyków 18, Poland.
| |
Collapse
|
2
|
Naidoo SJ, Naicker T. The Enigmatic Interplay of Interleukin-10 in the Synergy of HIV Infection Comorbid with Preeclampsia. Int J Mol Sci 2024; 25:9434. [PMID: 39273381 PMCID: PMC11395227 DOI: 10.3390/ijms25179434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Cytokines coordinate the intricate choreography of the immune system, directing cellular activities that mediate inflammation, pathogen defense, pathology and tissue repair. Within this spectrum, the anti-inflammatory prowess of interleukin-10 (IL-10) predominates in immune homeostasis. In normal pregnancy, the dynamic shift of IL-10 across trimesters maintains maternal immune tolerance ensuring fetal development and pregnancy success. Unravelling the dysregulation of IL-10 in pregnancy complications is vital, particularly in the heightened inflammatory condition of preeclampsia. Of note, a reduction in IL-10 levels contributes to endothelial dysfunction. In human immunodeficiency virus (HIV) infection, a complex interplay of IL-10 occurs, displaying a paradoxical paradigm of being immune-protective yet aiding viral persistence. Genetic variations in the IL-10 gene further modulate susceptibility to HIV infection and preeclampsia, albeit with nuanced effects across populations. This review outlines the conceptual framework underlying the role of IL-10 in the duality of normal pregnancy and preeclampsia together with HIV infection, thus highlighting its regulatory mechanisms and genetic influences. Synthesizing these findings in immune modulation presents avenues for therapeutic interventions in pregnancy complications comorbid with HIV infection.
Collapse
Affiliation(s)
| | - Thajasvarie Naicker
- Department of Optics and Imaging, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| |
Collapse
|
3
|
Hui R, Scheib CL, D’Atanasio E, Inskip SA, Cessford C, Biagini SA, Wohns AW, Ali MQ, Griffith SJ, Solnik A, Niinemäe H, Ge XJ, Rose AK, Beneker O, O’Connell TC, Robb JE, Kivisild T. Genetic history of Cambridgeshire before and after the Black Death. SCIENCE ADVANCES 2024; 10:eadi5903. [PMID: 38232165 PMCID: PMC10793959 DOI: 10.1126/sciadv.adi5903] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 12/14/2023] [Indexed: 01/19/2024]
Abstract
The extent of the devastation of the Black Death pandemic (1346-1353) on European populations is known from documentary sources and its bacterial source illuminated by studies of ancient pathogen DNA. What has remained less understood is the effect of the pandemic on human mobility and genetic diversity at the local scale. Here, we report 275 ancient genomes, including 109 with coverage >0.1×, from later medieval and postmedieval Cambridgeshire of individuals buried before and after the Black Death. Consistent with the function of the institutions, we found a lack of close relatives among the friars and the inmates of the hospital in contrast to their abundance in general urban and rural parish communities. While we detect long-term shifts in local genetic ancestry in Cambridgeshire, we find no evidence of major changes in genetic ancestry nor higher differentiation of immune loci between cohorts living before and after the Black Death.
Collapse
Affiliation(s)
- Ruoyun Hui
- Alan Turing Institute, London, UK
- McDonald Institute for Archaeological Research, University of Cambridge, Cambridge, UK
| | - Christiana L. Scheib
- McDonald Institute for Archaeological Research, University of Cambridge, Cambridge, UK
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia
- St John’s College, University of Cambridge, Cambridge, UK
| | | | - Sarah A. Inskip
- McDonald Institute for Archaeological Research, University of Cambridge, Cambridge, UK
- School of Archaeology and Ancient History, University of Leicester, Leicester, UK
| | - Craig Cessford
- McDonald Institute for Archaeological Research, University of Cambridge, Cambridge, UK
- Cambridge Archaeological Unit, Department of Archaeology, University of Cambridge, Cambridge, UK
| | | | - Anthony W. Wohns
- School of Medicine, Stanford University, Stanford, CA, USA
- Department of Genetics and Biology, Stanford University, Stanford, CA, USA
| | | | - Samuel J. Griffith
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anu Solnik
- Core Facility, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Helja Niinemäe
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Xiangyu Jack Ge
- Wellcome Genome Campus, Wellcome Sanger Institute, Hinxton, UK
| | - Alice K. Rose
- McDonald Institute for Archaeological Research, University of Cambridge, Cambridge, UK
- Department of Archaeology, University of Durham, Durham, UK
| | - Owyn Beneker
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Tamsin C. O’Connell
- McDonald Institute for Archaeological Research, University of Cambridge, Cambridge, UK
| | - John E. Robb
- Department of Archaeology, University of Cambridge, Cambridge, UK
| | - Toomas Kivisild
- McDonald Institute for Archaeological Research, University of Cambridge, Cambridge, UK
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
4
|
Redondo N, Rodríguez-Goncer I, Parra P, López-Medrano F, González E, Hernández A, Trujillo H, Ruiz-Merlo T, San Juan R, Folgueira MD, Andrés A, Aguado JM, Fernández-Ruiz M. Genetic polymorphisms in TLR3, IL10 and CD209 influence the risk of BK polyomavirus infection after kidney transplantation. Sci Rep 2022; 12:11338. [PMID: 35790769 PMCID: PMC9255529 DOI: 10.1038/s41598-022-15406-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 06/23/2022] [Indexed: 11/25/2022] Open
Abstract
Genetic determinants of BK polyomavirus infection after kidney transplantation remain poorly investigated. We assessed the potential impact of 13 different single nucleotide polymorphisms within genes mainly involved in innate immune responses on the risk of BKPyV viremia in 204 KT recipients. After a median follow-up of 1121.5 days, the cumulative incidence of any-level BKPyV viremia was 24.5% (50/204). There was a significant association between the minor T allele of TLR3 (rs3775291) SNP and the development of BKPyV viremia (adjusted hazard ratio [aHR]: 2.16; 95% confidence interval [CI]: 1.08–4.30; P value = 0.029), whereas the minor G allele of CD209 (rs4804803) SNP exerted a protective role (aHR: 0.54; 95% CI: 0.29–1.00; P value = 0.050). A higher incidence of BKPyV viremia was also observed for the minor G allele of IL10 (rs1800872) SNP, although the absence of BKPyV events among homozygotes for the reference allele prevented multivariable analysis. The BKPyV viremia-free survival rate decreased with the increasing number of unfavorable genotypes (100% [no unfavorable genotypes], 85.4% [1 genotype], 70.9% [2 genotypes], 52.5% [3 genotypes]; P value = 0.008). In conclusion, SNPs in TLR3, CD209 and IL10 genes play a role in modulating the susceptibility to any-level BKPyV viremia among KT recipients.
Collapse
Affiliation(s)
- Natalia Redondo
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Centro de Actividades Ambulatorias, 6ª planta, Bloque D. Avda. de Córdoba, s/n, 28041, Madrid, Spain. .,Centro de Investigación Biomédica en Red (CIBER) en Enfermedades Infecciosas, Madrid, Spain.
| | - Isabel Rodríguez-Goncer
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Centro de Actividades Ambulatorias, 6ª planta, Bloque D. Avda. de Córdoba, s/n, 28041, Madrid, Spain.,Centro de Investigación Biomédica en Red (CIBER) en Enfermedades Infecciosas, Madrid, Spain
| | - Patricia Parra
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Centro de Actividades Ambulatorias, 6ª planta, Bloque D. Avda. de Córdoba, s/n, 28041, Madrid, Spain.,Centro de Investigación Biomédica en Red (CIBER) en Enfermedades Infecciosas, Madrid, Spain
| | - Francisco López-Medrano
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Centro de Actividades Ambulatorias, 6ª planta, Bloque D. Avda. de Córdoba, s/n, 28041, Madrid, Spain.,Centro de Investigación Biomédica en Red (CIBER) en Enfermedades Infecciosas, Madrid, Spain.,Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Esther González
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Ana Hernández
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Hernando Trujillo
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Tamara Ruiz-Merlo
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Centro de Actividades Ambulatorias, 6ª planta, Bloque D. Avda. de Córdoba, s/n, 28041, Madrid, Spain.,Centro de Investigación Biomédica en Red (CIBER) en Enfermedades Infecciosas, Madrid, Spain
| | - Rafael San Juan
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Centro de Actividades Ambulatorias, 6ª planta, Bloque D. Avda. de Córdoba, s/n, 28041, Madrid, Spain.,Centro de Investigación Biomédica en Red (CIBER) en Enfermedades Infecciosas, Madrid, Spain.,Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| | - María Dolores Folgueira
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain.,Department of Microbiology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Amado Andrés
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain.,Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - José María Aguado
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Centro de Actividades Ambulatorias, 6ª planta, Bloque D. Avda. de Córdoba, s/n, 28041, Madrid, Spain.,Centro de Investigación Biomédica en Red (CIBER) en Enfermedades Infecciosas, Madrid, Spain.,Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Mario Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Centro de Actividades Ambulatorias, 6ª planta, Bloque D. Avda. de Córdoba, s/n, 28041, Madrid, Spain.,Centro de Investigación Biomédica en Red (CIBER) en Enfermedades Infecciosas, Madrid, Spain.,Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| |
Collapse
|
5
|
Dwivedi M, Laddha NC, Begum R. The Immunogenetics of Vitiligo: An Approach Toward Revealing the Secret of Depigmentation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:61-103. [PMID: 35286692 DOI: 10.1007/978-3-030-92616-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vitiligo is a hypomelanotic skin disease and considered to be of autoimmune origin due to breaching of immunological self-tolerance, resulting in inappropriate immune responses against melanocytes. The development of vitiligo includes a strong heritable component. Different strategies ranging from linkage studies to genome-wide association studies are used to explore the genetic factors responsible for the disease. Several vitiligo loci containing the respective genes have been identified which contribute to vitiligo and genetic variants for some of the genes are still unknown. These genes include mainly the proteins that play a role in immune regulation and a few other genes important for apoptosis and regulation of melanocyte functions. Despite the available data on genetic variants and risk alleles which influence the biological processes, only few immunological pathways have been found responsible for all ranges of severity and clinical manifestations of vitiligo. However, studies have concluded that vitiligo is of autoimmune origin and manifests due to complex interactions in immune components and their inappropriate response toward melanocytes. The genes involved in the immune regulation and processing the melanocytes antigen and its presentation can serve as effective immune-therapeutics that can target specific immunological pathways involved in vitiligo. This chapter highlights those immune-regulatory genes involved in vitiligo susceptibility and loci identified to date and their implications in vitiligo pathogenesis.
Collapse
Affiliation(s)
- Mitesh Dwivedi
- C. G. Bhakta Institute of Biotechnology, Uka Tarsadia University, Tarsadi, Surat, 394350, Gujarat, India.
| | - Naresh C Laddha
- In Vitro Specialty Lab Pvt. Ltd, 205-210, Golden Triangle, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Rasheedunnisa Begum
- Department of Biochemistry, The Maharaja Sayajirao University of Baroda, Vadodara, 390002, Gujarat, India
| |
Collapse
|
6
|
Further insight into the global variability of the OCA2-HERC2 locus for human pigmentation from multiallelic markers. Sci Rep 2021; 11:22530. [PMID: 34795370 PMCID: PMC8602267 DOI: 10.1038/s41598-021-01940-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/02/2021] [Indexed: 11/20/2022] Open
Abstract
The OCA2-HERC2 locus is responsible for the greatest proportion of eye color variation in humans. Numerous studies extensively described both functional SNPs and associated patterns of variation over this region. The goal of our study is to examine how these haplotype structures and allelic associations vary when highly variable markers such as microsatellites are used. Eleven microsatellites spanning 357 Kb of OCA2-HERC2 genes are analyzed in 3029 individuals from worldwide populations. We found that several markers display large differences in allele frequency (10% to 35% difference) among Europeans, East Asians and Africans. In Europe, the alleles showing increased frequency can also discriminate individuals with (IrisPlex) predicted blue and brown eyes. Distinct haplotypes are identified around the variants C and T of the functional SNP rs12913832 (associated to blue eyes), with linkage disequilibrium r2 values significant up to 237 Kb. The haplotype carrying the allele rs12913832 C has high frequency (76%) in blue eye predicted individuals (30% in brown eye predicted individuals), while the haplotype associated to the allele rs12913832 T is restricted to brown eye predicted individuals. Finally, homozygosity values reach levels of 91% near rs12913832. Odds ratios show values of 4.2, 7.4 and 10.4 for four markers around rs12913832 and 7.1 for their core haplotype. Hence, this study provides an example on the informativeness of multiallelic markers that, despite their current limited potential contribution to forensic eye color prediction, supports the use of microsatellites for identifying causing variants showing similar genetic features and history.
Collapse
|
7
|
Dabitao D, Dembele M, Urbanowski M, Kone B, Wague M, Coulibaly N, Sarro YDS, Baya B, Goita D, Dao S, Belson M, Klein SL, Achenbach C, Holl JL, Diakite M, Doumbia S, Bream JH, Bishai WR, Diallo S, Murphy RL. Short Communication: Genetic Variation in Human IL10 Proximal Promoter and Susceptibility to HIV-1 Infection in Mali, West Africa. AIDS Res Hum Retroviruses 2021; 37:57-61. [PMID: 33045845 DOI: 10.1089/aid.2020.0140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It is now recognized that to fully understand the role of host genetic variation on susceptibility to HIV-1 infection, investigations must be extended to African populations. We sought to determine if genetic variation in IL10 are associated with HIV-1 infection in a West African cohort in Mali. HIV-infected and -uninfected individuals were genotyped for three common single nucleotide polymorphisms (SNPs) located at positions -592 (C/A), -819 (C/T), and -1082 (G/A) of the IL10 promoter. We found that the ATA haplotype, which has been previously associated with low IL-10 expression, was the most represented in the cohort. Although we observed a trend toward an increased frequency of ATA/ATA carriage in HIV-infected compared with -uninfected individuals, the difference was not statistically significant. Similarly, individual IL10 SNPs were not significantly enriched in the HIV-infected group, suggesting that IL10 genetic variants are not associated with HIV-1 in this West African cohort from Mali.
Collapse
Affiliation(s)
- Djeneba Dabitao
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
- School for Professional Studies, Northwestern University (NU), Chicago, Illinois, USA
| | - Mamadou Dembele
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
| | - Michael Urbanowski
- Department of Infectious Diseases, Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Bourahima Kone
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
| | - Mamadou Wague
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
| | - Nadie Coulibaly
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
| | - Yeya dit Sadio Sarro
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
| | - Bocar Baya
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
| | - Drissa Goita
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
| | - Sounkalo Dao
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
| | - Michael Belson
- Collaborative Clinical Research Branch (CCRB), Division of Clinical Research (DCR), National Institutes of Allergy and Infectious Diseases (NIAID), Bethesda, Maryland, USA
| | - Sabra L. Klein
- Johns Hopkins Bloomberg School of Public Health, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Baltimore, Maryland, USA
| | - Chad Achenbach
- Division of Infectious Diseases and Institute for Global Health, Feinberg School of Medicine, Northwestern University (NU), Chicago, Illinois, USA
- Biological Sciences Division, University of Chicago, Chicago, Illinois, USA
| | - Jane L. Holl
- Division of Infectious Diseases and Institute for Global Health, Feinberg School of Medicine, Northwestern University (NU), Chicago, Illinois, USA
- Biological Sciences Division, University of Chicago, Chicago, Illinois, USA
| | - Mahamadou Diakite
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
| | - Seydou Doumbia
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
| | - Jay H. Bream
- Johns Hopkins Bloomberg School of Public Health, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Baltimore, Maryland, USA
- Graduate Program in Immunology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - William R. Bishai
- Department of Infectious Diseases, Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Souleymane Diallo
- Faculty of Pharmacy and Faculty of Medicine and Odonto-Stomatology, University Clinical Research Center (UCRC), University of Sciences, Techniques, and Technologies of Bamako (USTTB), Mali, West Africa
| | - Robert L. Murphy
- Division of Infectious Diseases and Institute for Global Health, Feinberg School of Medicine, Northwestern University (NU), Chicago, Illinois, USA
- Biological Sciences Division, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
8
|
Miller H, Czigany Z, Lurje I, Reichelt S, Bednarsch J, Strnad P, Trautwein C, Roderburg C, Tacke F, Gaisa NT, Knüchel-Clarke R, Neumann UP, Lurje G. Impact of Angiogenesis- and Hypoxia-Associated Polymorphisms on Tumor Recurrence in Patients with Hepatocellular Carcinoma Undergoing Surgical Resection. Cancers (Basel) 2020; 12:cancers12123826. [PMID: 33352897 PMCID: PMC7767259 DOI: 10.3390/cancers12123826] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/13/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Hepatocellular carcinoma remains a leading cause of cancer-related death and the most common primary hepatic malignancy in the Western hemisphere. Previous research found that angiogenesis-related cytokines and elevated levels of interleukin 8 and vascular endothelial growth factor (VEGF) shorten the expected time of survival. Moreover, factors of tumor angiogenesis- and hypoxia-driven signaling pathways are already associated with worse outcome in disease-free survival in several tumor entities. Our study investigates the prognosis of hepatocellular carcinoma patients based on a selection of ten different single-nucleotide polymorphisms from angiogenesis, carcinogenesis, and hypoxia pathways. Our study with 127 patients found supporting evidence that polymorphisms in angiogenesis-associated pathways corelate with disease-free survival and clinical outcome in patients with hepatocellular carcinoma. Abstract Tumor angiogenesis plays a pivotal role in hepatocellular carcinoma (HCC) biology. Identifying molecular prognostic markers is critical to further improve treatment selection in these patients. The present study analyzed a subset of 10 germline polymorphisms involved in tumor angiogenesis pathways and their impact on prognosis in HCC patients undergoing partial hepatectomy in a curative intent. Formalin-fixed paraffin-embedded (FFPE) tissues were obtained from 127 HCC patients at a German primary care hospital. Genomic DNA was extracted, and genotyping was carried out using polymerase chain reaction (PCR)–restriction fragment length polymorphism-based protocols. Polymorphisms in interleukin-8 (IL-8) (rs4073; p = 0.047, log-rank test) and vascular endothelial growth factor (VEGF C + 936T) (rs3025039; p = 0.045, log-rank test) were significantly associated with disease-free survival (DFS). After adjusting for covariates in the multivariable model, IL-8 T-251A (rs4073) (adjusted p = 0.010) and a combination of “high-expression” variants of rs4073 and rs3025039 (adjusted p = 0.034) remained significantly associated with DFS. High-expression variants of IL-8 T-251A may serve as an independent molecular marker of prognosis in patients undergoing surgical resection for HCC. Assessment of the patients’ individual genetic risks may help to identify patient subgroups at high risk for recurrence following curative-intent surgery.
Collapse
Affiliation(s)
- Hannah Miller
- Charité–Universitätsmedizin Berlin, Department of Surgery, Campus Charité Mitte|Campus Virchow-Klinikum, 13353 Berlin, Germany; (H.M.); (S.R.)
- Department of Surgery and Transplantation, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (Z.C.); (I.L.); (J.B.); (U.P.N.)
| | - Zoltan Czigany
- Department of Surgery and Transplantation, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (Z.C.); (I.L.); (J.B.); (U.P.N.)
| | - Isabella Lurje
- Department of Surgery and Transplantation, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (Z.C.); (I.L.); (J.B.); (U.P.N.)
- Charité–Universitätsmedizin Berlin, Department of Gastroenterology and Hepatology, Campus Charité Mitte|Campus Virchow-Klinikum, 13353 Berlin, Germany; (C.R.); (F.T.)
| | - Sophie Reichelt
- Charité–Universitätsmedizin Berlin, Department of Surgery, Campus Charité Mitte|Campus Virchow-Klinikum, 13353 Berlin, Germany; (H.M.); (S.R.)
- Department of Surgery and Transplantation, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (Z.C.); (I.L.); (J.B.); (U.P.N.)
| | - Jan Bednarsch
- Department of Surgery and Transplantation, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (Z.C.); (I.L.); (J.B.); (U.P.N.)
| | - Pavel Strnad
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (P.S.); (C.T.)
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (P.S.); (C.T.)
| | - Christoph Roderburg
- Charité–Universitätsmedizin Berlin, Department of Gastroenterology and Hepatology, Campus Charité Mitte|Campus Virchow-Klinikum, 13353 Berlin, Germany; (C.R.); (F.T.)
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (P.S.); (C.T.)
| | - Frank Tacke
- Charité–Universitätsmedizin Berlin, Department of Gastroenterology and Hepatology, Campus Charité Mitte|Campus Virchow-Klinikum, 13353 Berlin, Germany; (C.R.); (F.T.)
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (P.S.); (C.T.)
| | - Nadine Therese Gaisa
- Institute of Pathology, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (N.T.G.); (R.K.-C.)
| | - Ruth Knüchel-Clarke
- Institute of Pathology, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (N.T.G.); (R.K.-C.)
| | - Ulf Peter Neumann
- Department of Surgery and Transplantation, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (Z.C.); (I.L.); (J.B.); (U.P.N.)
| | - Georg Lurje
- Charité–Universitätsmedizin Berlin, Department of Surgery, Campus Charité Mitte|Campus Virchow-Klinikum, 13353 Berlin, Germany; (H.M.); (S.R.)
- Department of Surgery and Transplantation, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, 52074 Aachen, Germany; (Z.C.); (I.L.); (J.B.); (U.P.N.)
- Correspondence: ; Tel.: +49-30-450-652339
| |
Collapse
|
9
|
Chetty A, Omondi MA, Butters C, Smith KA, Katawa G, Ritter M, Layland L, Horsnell W. Impact of Helminth Infections on Female Reproductive Health and Associated Diseases. Front Immunol 2020; 11:577516. [PMID: 33329545 PMCID: PMC7719634 DOI: 10.3389/fimmu.2020.577516] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/27/2020] [Indexed: 12/25/2022] Open
Abstract
A growing body of knowledge exists on the influence of helminth infections on allergies and unrelated infections in the lung and gastrointestinal (GI) mucosa. However, the bystander effects of helminth infections on the female genital mucosa and reproductive health is understudied but important considering the high prevalence of helminth exposure and sexually transmitted infections in low- and middle-income countries (LMICs). In this review, we explore current knowledge about the direct and systemic effects of helminth infections on unrelated diseases. We summarize host disease-controlling immunity of important sexually transmitted infections and introduce the limited knowledge of how helminths infections directly cause pathology to female reproductive tract (FRT), alter susceptibility to sexually transmitted infections and reproduction. We also review work by others on type 2 immunity in the FRT and hypothesize how these insights may guide future work to help understand how helminths alter FRT health.
Collapse
Affiliation(s)
- Alisha Chetty
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Millicent A Omondi
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Claire Butters
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Katherine Ann Smith
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa.,School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Gnatoulma Katawa
- Ecole Supérieure des Techniques Biologiques et Alimentaires, Université de Lomé, Lomé, Togo
| | - Manuel Ritter
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
| | - Laura Layland
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
| | - William Horsnell
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa.,Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
10
|
Tamalet C, Devaux C, Dubourg G, Colson P. Resistance to human immunodeficiency virus infection: a rare but neglected state. Ann N Y Acad Sci 2020; 1485:22-42. [PMID: 33009659 DOI: 10.1111/nyas.14452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/25/2020] [Accepted: 07/07/2020] [Indexed: 11/29/2022]
Abstract
The natural history of human immunodeficiency virus (HIV) infection is well understood. In most individuals sexually exposed to HIV, the risk of becoming infected depends on the viral load and on sexual practices and gender. However, a low percentage of individuals who practice frequent unprotected sexual intercourse with HIV-infected partners remain uninfected. Although the systematic study of these individuals has made it possible to identify HIV resistance factors including protective genetic patterns, such epidemiological situations remain paradoxical and not fully understood. In vitro experiments have demonstrated that peripheral blood mononuclear cells (PBMCs) from HIV-free, unexposed blood donors are not equally susceptible to HIV infection; in addition, PBMCs from highly exposed seronegative individuals are generally resistant to infection by primary HIV clinical isolates. We review the literature on permissiveness of PBMCs from healthy blood donors and uninfected hyperexposed individuals to sustained infection and replication of HIV-1 in vitro. In addition, we focus on recent evidence indicating that the gut microbiota may either contribute to natural resistance to or delay replication of HIV infected individuals.
Collapse
Affiliation(s)
- Catherine Tamalet
- IHU Méditerranée Infection and Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| | - Christian Devaux
- IHU Méditerranée Infection and Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| | - Gregory Dubourg
- IHU Méditerranée Infection and Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| | - Philippe Colson
- IHU Méditerranée Infection and Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| |
Collapse
|
11
|
Thami PK, Chimusa ER. Population Structure and Implications on the Genetic Architecture of HIV-1 Phenotypes Within Southern Africa. Front Genet 2019; 10:905. [PMID: 31611910 PMCID: PMC6777512 DOI: 10.3389/fgene.2019.00905] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022] Open
Abstract
The interesting history of Southern Africa has put the region in the spotlight for population medical genetics. Major events including the Bantu expansion and European colonialism have imprinted unique genetic signatures within autochthonous populations of Southern Africa, this resulting in differential allele frequencies across the region. This genetic structure has potential implications on susceptibility and resistance to infectious diseases such as human immunodeficiency virus (HIV) infection. Southern Africa is the region affected worst by HIV. Here, we discuss advances made in genome-wide association studies (GWAS) of HIV-1 in the past 12 years and dissect population diversity within Southern Africa. Our findings accentuate that a plethora of factors such as migration, language and culture, admixture, and natural selection have profiled the genetics of the people of Southern Africa. Genetic structure has been observed among the Khoe-San, among Bantu speakers, and between the Khoe-San, Coloureds, and Bantu speakers. Moreover, Southern African populations have complex admixture scenarios. Few GWAS of HIV-1 have been conducted in Southern Africa, with only one of these identifying two novel variants (HCG22rs2535307 and CCNG1kgp22385164) significantly associated with HIV-1 acquisition and progression. High genetic diversity, multi-wave genetic mixture and low linkage disequilibrium of Southern African populations constitute a challenge in identifying genetic variants with modest risk or protective effect against HIV-1. We therefore posit that it is compelling to assess genome-wide contribution of ancestry to HIV-1 infection. We further suggest robust methods that can pin-point population-specific variants that may contribute to the control of HIV-1 in Southern Africa.
Collapse
Affiliation(s)
- Prisca K Thami
- Division of Human Genetics, Department of Pathology, University of Cape Town, Cape Town, South Africa.,Research Laboratory, Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Emile R Chimusa
- Division of Human Genetics, Department of Pathology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
12
|
Kannagi M, Hasegawa A, Nagano Y, Kimpara S, Suehiro Y. Impact of host immunity on HTLV-1 pathogenesis: potential of Tax-targeted immunotherapy against ATL. Retrovirology 2019; 16:23. [PMID: 31438973 PMCID: PMC6704564 DOI: 10.1186/s12977-019-0484-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/12/2019] [Indexed: 12/18/2022] Open
Abstract
Human T-cell leukemia virus type-1 (HTLV-1) causes adult T-cell leukemia/lymphoma (ATL), HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), and other inflammatory diseases. There is no disease-specific difference in viral strains, and it is unclear how HTLV-1 causes such different diseases manifesting as lymphoproliferation or inflammation. Although some progress has been made in therapies for these diseases, the prognosis for ATL is still dismal and HAM/TSP remains an intractable disease. So far, two regulatory proteins of HTLV-1, Tax and HBZ, have been well studied and shown to have pleiotropic functions implicated in viral pathogenesis. Tax in particular can strongly activate NFκB, which is constitutively activated in HTLV-1-infected cells and considered to contribute to both oncogenesis and inflammation. However, the expression level of Tax is very low in vivo, leading to confusion in understanding its role in viral pathogenesis. A series of studies using IL-2-dependent HTLV-1-infected cells indicated that IL-10, an anti-inflammatory/immune suppressive cytokine, could induce a proliferative phenotype in HTLV-1-infected cells. In addition, type I interferon (IFN) suppresses HTLV-1 expression in a reversible manner. These findings suggest involvement of host innate immunity in the switch between lymphoproliferative and inflammatory diseases as well as the regulation of HTLV-1 expression. Innate immune responses also affect another important host determinant, Tax-specific cytotoxic T lymphocytes (CTLs), which are impaired in ATL patients, while activated in HAM/TSP patients. Activation of Tax-specific CTLs in ATL patients after hematopoietic stem cell transplantation indicates Tax expression and its fluctuation in vivo. A recently developed anti-ATL therapeutic vaccine, consisting of Tax peptide-pulsed dendritic cells, induced Tax-specific CTL responses in ATL patients and exhibited favorable clinical outcomes, unless Tax-defective ATL clones emerged. These findings support the significance of Tax in HTLV-1 pathogenesis, at least in part, and encourage Tax-targeted immunotherapy in ATL. Host innate and acquired immune responses induce host microenvironments that modify HTLV-1-encoded pathogenesis and establish a complicated network for development of diseases in HTLV-1 infection. Both host and viral factors should be taken into consideration in development of therapeutic and prophylactic strategies in HTLV-1 infection.
Collapse
Affiliation(s)
- Mari Kannagi
- Department of Immunotherapeutics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.
| | - Atsuhiko Hasegawa
- Department of Immunotherapeutics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Yoshiko Nagano
- Department of Immunotherapeutics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Shuichi Kimpara
- Department of Immunotherapeutics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.,Department of Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Youko Suehiro
- Department of Hematology, National Kyushu Cancer Center, Fukuoka, Japan
| |
Collapse
|
13
|
Ramírez de Arellano E, Díez-Fuertes F, Aguilar F, de la Torre Tarazona HE, Sánchez-Lara S, Lao Y, Vicario JL, García F, González-Garcia J, Pulido F, Gutierrez-Rodero F, Moreno S, Iribarren JA, Viciana P, Vilches C, Ramos M, Capa L, Alcamí J, Del Val M. Novel association of five HLA alleles with HIV-1 progression in Spanish long-term non progressor patients. PLoS One 2019; 14:e0220459. [PMID: 31393887 PMCID: PMC6687284 DOI: 10.1371/journal.pone.0220459] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 07/16/2019] [Indexed: 12/12/2022] Open
Abstract
Certain host genetic variants, especially in the human leucocyte antigen (HLA) region, are associated with different progression of HIV-1-induced diseases and AIDS. Long term non progressors (LTNP) represent only the 2% of infected patients but are especially relevant because of their efficient HIV control. In this work we present a global analysis of genetic data in the large national multicenter cohort of Spanish LTNP, which is compared with seronegative individuals and HIV-positive patients. We have analyzed whether several single-nucleotide polymorphisms (SNPs) including in key genes and certain HLA-A and B alleles could be associated with a specific HIV phenotype. A total of 846 individuals, 398 HIV-1-positive patients (213 typical progressors, 55 AIDS patients, and 130 LTNPs) and 448 HIV-negative controls, were genotyped for 15 polymorphisms and HLA-A and B alleles. Significant differences in the allele frequencies among the studied populations identified 16 LTNP-associated genetic factors, 5 of which were defined for the first time as related to LTNP phenotype: the protective effect of HLA-B39, and the detrimental impact of HLA-B18, -A24, -B08 and –A29. The remaining eleven polymorphisms confirmed previous publications, including the protective alleles HLA-B57, rs2395029 (HCP5), HLA bw4 homozygosity, HLA-B52, HLA-B27, CCR2 V64I, rs9264942 (HLA-C) and HLA-A03; and the risk allele HLA bw6 homozygosity. Notably, individual Spanish HIV-negative individuals had an average of 0.12 protective HLA alleles and SNPs, compared with an average of 1.43 protective alleles per LTNP patient, strongly suggesting positive selection of LTNP. Finally, stratification of LTNP according to viral load showed a proportional relationship between the frequency of protective alleles with control of viral load. Interestingly, no differences in the frequency of protection/risk polymorphisms were found between elite controllers and LTNPs maintaining viral loads <2.000 copies/mL throughout the follow-up.
Collapse
Affiliation(s)
- Eva Ramírez de Arellano
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- * E-mail:
| | - Francisco Díez-Fuertes
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Infectious Diseases Unit, IBIDAPS, HIVACAT, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Francisco Aguilar
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | | | - Susana Sánchez-Lara
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Viral Immunology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Yolanda Lao
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - José Luis Vicario
- Departamento de Histocompatibilidad, Centro de Transfusión de Madrid, Madrid, Spain
| | - Felipe García
- Infectious Diseases Unit, IBIDAPS, HIVACAT, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | | | - Federico Pulido
- HIV Unit, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Félix Gutierrez-Rodero
- Servicio de Medicina Interna, Unidad de Enfermedades Infecciosas, Hospital General Universitario de Elche, Alicante, Spain
| | | | | | - Pompeyo Viciana
- Laboratory of Immunovirology, Biomedicine Institute of Sevilla, Virgen del Rocío University Hospital, Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, IBIS/CSIC/SAS/University of Sevilla, Sevilla, Spain
| | - Carlos Vilches
- Inmunogenética e Histocompatibilidad, Instituto de Investigación Sanitaria Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Manuel Ramos
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Viral Immunology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Laura Capa
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - José Alcamí
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Infectious Diseases Unit, IBIDAPS, HIVACAT, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Margarita Del Val
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Viral Immunology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| |
Collapse
|
14
|
Singh P, Rajput R, Mehra NK, Vajpayee M, Sarin R. Cytokine gene polymorphisms among North Indians: Implications for genetic predisposition? INFECTION GENETICS AND EVOLUTION 2019; 73:450-459. [PMID: 31173933 DOI: 10.1016/j.meegid.2019.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/01/2019] [Accepted: 06/03/2019] [Indexed: 11/25/2022]
Abstract
Variations in the production and activity of cytokines influence the susceptibility and/or resistance to various infectious agents, autoimmune diseases, as well as the post-transplant engraftment/ rejection. Differences in the production of cytokines between individuals have been correlated to single nucleotide polymorphisms (SNPs) in the promoter, coding or non-coding regions of cytokine genes. The present study aimed at understanding distribution of cytokine gene variants among HIV seropositive subjects including HIV + TB+ subjects of Indian origin. Our findings indicate significant association of pro-inflammatory (IL2, IFN-γ, TNF-α) and anti-inflammatory cytokine gene variants (IL4, IL10) with the risk to acquire the HIV infection and development of AIDS related illness in Indian population. Since distribution of genetic polymorphisms varies significantly across different populations, different genotypes might exhibit different disease-modifying effects. An understanding of the immunogenetic factors or AIDS restriction genes is important not only for elucidating the mechanisms of disease pathogenesis but also for vaccine design and its application.
Collapse
Affiliation(s)
- Paras Singh
- Department of Molecular Medicine, National Institute of Tuberculosis and Respiratory Diseases, Sri Aurobindo Marg, New Delhi, India.
| | - Roopali Rajput
- Department of Molecular Medicine, National Institute of Tuberculosis and Respiratory Diseases, Sri Aurobindo Marg, New Delhi, India
| | - Narinder K Mehra
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Madhu Vajpayee
- Department of Microbiology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Rohit Sarin
- Department of TB and Respiratory Diseases, National Institute of Tuberculosis and Respiratory Diseases, Sri Aurobindo Marg, New Delhi, India
| |
Collapse
|
15
|
Singh H, Samani D, Nain S, Dhole TN. Interleukin-10 polymorphisms and susceptibility to ARV associated hepatotoxicity. Microb Pathog 2019; 133:103544. [PMID: 31121270 DOI: 10.1016/j.micpath.2019.103544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 05/11/2019] [Accepted: 05/17/2019] [Indexed: 12/26/2022]
Abstract
Interleukin-10 (IL-10) is an anti-inflammatory cytokine associated with the inhibition of HIV replication. IL-10 polymorphisms were found to be linked to drug-induced hepatotoxicity. Hence we examined the prevalence of IL-10 (-819C/T,-1082A/G) polymorphisms in a total of 165 HIV patients which included 34 patients with hepatotoxicity, 131 without hepatotoxicity and 155 healthy controls by the PCR-RFLP method. In HIV patients with hepatotoxicity, the IL-10-819TT genotype increased the risk of ARV associated hepatotoxicity severity (OR = 1.61, P = 0.35). IL-10-819TT genotype was overrepresented in patients with hepatotoxicity as compared to healthy controls (26.5% vs. 13.5%, OR = 1.61, P = 0.46). IL-10 -819CT genotype was associated with advance HIV disease stage (OR = 0.49, P = 0.045). In HIV patients without hepatotoxicity, the IL-10-819TT genotype was more prevalent in patients consuming tobacco as compared to non-users (OR = 1.60, P = 0.41). In HIV patients without hepatotoxicity using both alcohol + efavirenz along with IL-10 -819CT genotype resulted in increased risk for the acquisition of ARV associated hepatotoxicity (OR = 4.00, P = 0.36). In multivariate logistic regression, taking nevirapine was associated with the risk hepatotoxicity severity (OR = 0.23, P = 0.005). In conclusion, an insignificant association between IL-10 polymorphisms and susceptibility to ARV associated hepatotoxicity.
Collapse
Affiliation(s)
- HariOm Singh
- Department of Molecular Biology, National AIDS Research Institute Pune, 411026, India.
| | - Dharmesh Samani
- Department of Molecular Biology, National AIDS Research Institute Pune, 411026, India
| | - Sumitra Nain
- Department of Pharmacy, University of Banasthali, Banasthali Vidyapith, Jaipur, 302001, India
| | - T N Dhole
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, 226014, India
| |
Collapse
|
16
|
Harishankar M, Ravikrishnan H, Ravishankar A, Hanna LE, Swaminathan S, Selvaraj P, Bethunaickan R. IL-10 Promoter -592 Polymorphism may Influence Susceptibility to HIV Infection in South Indian Population. Curr HIV Res 2019; 16:58-63. [PMID: 29468971 DOI: 10.2174/1570162x16666180219153752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/02/2018] [Accepted: 02/12/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Genetic factors play an important role in the development of disease susceptibility or protection. Cytokine gene polymorphisms are reported to be associated with altered levels of cytokine production that can impact disease progression in HIV and TB. OBJECTIVE In this study, we studied IL-10 -592(C/A) and TGF-β -509 (C/T) promoter polymorphisms to understand their role in susceptibility or resistance to HIV and TB in a South Indian population. METHOD Genomic DNA was isolated from healthy controls, pulmonary tuberculosis patients (n=122) and HIV positive individuals (n=100) and used for genotyping by polymerase chain reaction followed by restriction fragment length polymorphism (PCR-RFLP) method. RESULTS Results revealed that under dominant model (CC vs CA+AA), IL-10 -592 'A' allele either 'CA' or 'AA' combinations significantly associated with susceptibility to HIV compared to healthy controls (OR: 1.88(1.05-3.35); p=0.030). However, we found no significant association with TB. TGF-β -509 polymorphism did not associate with either HIV or TB under overdominant model. Neither of the promoter polymorphisms associated with sex in either HIV or TB. However, a trend towards higher risk to HIV was found in females compared with males in IL-10 -592 'AA' genotype. CONCLUSION This study suggests the association of IL-10 -592 "AA" genotype with susceptibility to HIV under dominant model in the Southern Indian population. Future studies are needed with a larger sample size in order to confirm the observations made in this study.
Collapse
Affiliation(s)
- Murugesan Harishankar
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai, India
| | - Harini Ravikrishnan
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai, India
| | - Akshaya Ravishankar
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai, India
| | - Luke Elizabeth Hanna
- Division of HIV, Department of Clinical Research, National Institute for Research in Tuberculosis, Chennai, India
| | - Soumya Swaminathan
- Division of HIV, Department of Clinical Research, National Institute for Research in Tuberculosis, Chennai, India.,Director- General, Indian Council of Medical Research, New Delhi, India
| | - Paramasivam Selvaraj
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai, India
| | | |
Collapse
|
17
|
Roider J, Ngoepe A, Muenchhoff M, Adland E, Groll A, Ndung'u T, Kløverpris H, Goulder P, Leslie A. Increased Regulatory T-Cell Activity and Enhanced T-Cell Homeostatic Signaling in Slow Progressing HIV-infected Children. Front Immunol 2019; 10:213. [PMID: 30809229 PMCID: PMC6379343 DOI: 10.3389/fimmu.2019.00213] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/24/2019] [Indexed: 12/14/2022] Open
Abstract
Pediatric slow progressors (PSP) are rare ART-naïve, HIV-infected children who maintain high CD4 T-cell counts and low immune activation despite persistently high viral loads. Using a well-defined cohort of PSP, we investigated the role of regulatory T-cells (TREG) and of IL-7 homeostatic signaling in maintaining normal-for-age CD4 counts in these individuals. Compared to children with progressive disease, PSP had greater absolute numbers of TREG, skewed toward functionally suppressive phenotypes. As with immune activation, overall T-cell proliferation was lower in PSP, but was uniquely higher in central memory TREG (CM TREG), indicating active engagement of this subset. Furthermore, PSP secreted higher levels of the immunosuppressive cytokine IL-10 than children who progressed. The frequency of suppressive TREG, CM TREG proliferation, and IL-10 production were all lower in PSP who go on to progress at a later time-point, supporting the importance of an active TREG response in preventing disease progression. In addition, we find that IL-7 homeostatic signaling is enhanced in PSP, both through preserved surface IL-7receptor (CD127) expression on central memory T-cells and increased plasma levels of soluble IL-7receptor, which enhances the bioactivity of IL-7. Combined analysis, using a LASSO modeling approach, indicates that both TREG activity and homeostatic T-cell signaling make independent contributions to the preservation of CD4 T-cells in HIV-infected children. Together, these data demonstrate that maintenance of normal-for-age CD4 counts in PSP is an active process, which requires both suppression of immune activation through functional TREG, and enhanced T-cell homeostatic signaling.
Collapse
Affiliation(s)
- Julia Roider
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, Oxford University, Oxford, United Kingdom
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Infectious Diseases, Medizinische Klinik IV, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Abigail Ngoepe
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Maximilian Muenchhoff
- Department of Virology, Max von Pettenkofer Institute, Ludwig-Maximilians-University Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Emily Adland
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, Oxford University, Oxford, United Kingdom
| | - Andreas Groll
- Faculty of Statistics, TU Dortmund University, Dortmund, Germany
| | - Thumbi Ndung'u
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
- Max Planck Institute for Infection Biology, Berlin, Germany
- Department of Infection and Immunity, University College London, London, United Kingdom
| | - Henrik Kløverpris
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Infection and Immunity, University College London, London, United Kingdom
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Philip Goulder
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, Oxford University, Oxford, United Kingdom
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Alasdair Leslie
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
18
|
Abstract
Exhausted CD8 T (Tex) cells are a distinct cell lineage that arise during chronic infections and cancers in animal models and humans. Tex cells are characterized by progressive loss of effector functions, high and sustained inhibitory receptor expression, metabolic dysregulation, poor memory recall and homeostatic self-renewal, and distinct transcriptional and epigenetic programs. The ability to reinvigorate Tex cells through inhibitory receptor blockade, such as αPD-1, highlights the therapeutic potential of targeting this population. Emerging insights into the mechanisms of exhaustion are informing immunotherapies for cancer and chronic infections. However, like other immune cells, Tex cells are heterogeneous and include progenitor and terminal subsets with unique characteristics and responses to checkpoint blockade. Here, we review our current understanding of Tex cell biology, including the developmental paths, transcriptional and epigenetic features, and cell intrinsic and extrinsic factors contributing to exhaustion and how this knowledge may inform therapeutic targeting of Tex cells in chronic infections, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Laura M McLane
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; .,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Mohamed S Abdel-Hakeem
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; .,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo 11562, Egypt
| | - E John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; .,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
19
|
Kim IS, Kim HH, Chang CL. Genetic Variants and Haplotypes in the IL10 Gene and Their Association with Opportunistic Infections among HIV-Infected Patients in Korea in the Era of Highly Active Antiretroviral Therapy. ANNALS OF CLINICAL MICROBIOLOGY 2019. [DOI: 10.5145/acm.2019.22.1.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- In-Suk Kim
- Department of Laboratory Medicine and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Busan, Korea
| | - Hyung-Hoi Kim
- Department of Laboratory Medicine and Biomedical Research Institute, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea
| | - Chulhun L. Chang
- Department of Laboratory Medicine and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Busan, Korea
| |
Collapse
|
20
|
|
21
|
Tsiara CG, Nikolopoulos GK, Dimou NL, Pantavou KG, Bagos PG, Mensah B, Talias M, Braliou GG, Paraskeva D, Bonovas S, Hatzakis A. Interleukin gene polymorphisms and susceptibility to HIV-1 infection: a meta-analysis. J Genet 2018; 97:235-251. [PMID: 29666343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Some subjects are repeatedly exposed to human immunodeficiency virus (HIV), yet they remain uninfected. This suggests the existence of host-resistance mechanisms. The current study synthesizes the evidence regarding the association between interleukin (IL) gene polymorphisms and HIV susceptibility. Medline, Scopus and the Web of Science databases were systematically searched, and a meta-analysis of case-control studies was conducted. Univariate and bivariate methods were used. The literature search identified 42 eligible studies involving 15,727 subjects. Evidence was obtained on eight single-nucleotide polymorphisms (SNPs): IL1A -889 C>T (rs1800587), IL1B +3953/4 C>T (rs1143634), IL4 -589/90 C>T (rs2243250), IL6 -174 G>C (rs1800795), IL10 -592 C>A (rs1800872), IL10-1082 A>G (rs1800896), IL12B -1188 A>C (rs3212227) and IL28B C>T (rs12979860). The IL1B +3953/4 C>T variant appears to increase the risk of HIV acquisition, under the assumption of a recessive genetic model (odds ratio (OR): 4.47, 95% CI: 2.35-8.52). The AA homozygotes of the IL10 -592 C>A SNP had an increased, marginally nonsignificant, risk (OR: 1.39, 95% CI: 0.97-2.01). It reached, however, significance in sub analyses (OR: 1.49, 95% CI: 1.04-2.12). Finally, the well-studied hepatitis C virus (HCV) infection IL28B (rs12979860) CT/TT genotypes were associated with a 27% decrease in HIV infection risk, especially in populations infected with HCV (OR: 0.73, 95% CI: 0.57-0.95). Interleukin signalling is perhaps important in HIV infection and some interleukin genetic variants may affect the risk of HIV acquisition. Approaches targeting specific genes and genome wide association studies should be conducted to decipher the effect of these polymorphisms.
Collapse
Affiliation(s)
- Chrissa G Tsiara
- Hellenic Centre for Disease Control and Prevention, 15123 Athens, Greece. ,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Hofmann SR, Kapplusch F, Mäbert K, Hedrich CM. The molecular pathophysiology of chronic non-bacterial osteomyelitis (CNO)-a systematic review. Mol Cell Pediatr 2017; 4:7. [PMID: 28685269 PMCID: PMC5500598 DOI: 10.1186/s40348-017-0073-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/27/2017] [Indexed: 02/07/2023] Open
Abstract
Chronic non-bacterial osteomyelitis (CNO) belongs to the growing spectrum of autoinflammatory diseases and primarily affects the skeletal system. Peak onset ranges between 7 and 12 years of age. The clinical spectrum of CNO covers sometimes asymptomatic inflammation of single bones at the one end and chronically active or recurrent multifocal osteitis at the other.Despite the intense scientific efforts, the exact molecular mechanisms of CNO remain unknown. Recent data suggest CNO as a genetically complex disorder with dysregulated TLR4/MAPK/inflammasome signaling cascades resulting in an imbalance between pro- and anti-inflammatory cytokine expression, leading to osteoclast activation and osteolytic lesions.In this manuscript, the current understanding of molecular patho-mechanisms in CNO will be discussed.
Collapse
Affiliation(s)
- Sigrun Ruth Hofmann
- Pediatric Rheumatology and Immunology, Children's Hospital Dresden, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, D-01307, Dresden, Germany
| | - Franz Kapplusch
- Pediatric Rheumatology and Immunology, Children's Hospital Dresden, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, D-01307, Dresden, Germany
| | - Katrin Mäbert
- Pediatric Rheumatology and Immunology, Children's Hospital Dresden, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, D-01307, Dresden, Germany
| | - Christian Michael Hedrich
- Pediatric Rheumatology and Immunology, Children's Hospital Dresden, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, D-01307, Dresden, Germany.
- Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
- Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK.
| |
Collapse
|
23
|
Sawada L, Nagano Y, Hasegawa A, Kanai H, Nogami K, Ito S, Sato T, Yamano Y, Tanaka Y, Masuda T, Kannagi M. IL-10-mediated signals act as a switch for lymphoproliferation in Human T-cell leukemia virus type-1 infection by activating the STAT3 and IRF4 pathways. PLoS Pathog 2017; 13:e1006597. [PMID: 28910419 PMCID: PMC5614654 DOI: 10.1371/journal.ppat.1006597] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/26/2017] [Accepted: 08/22/2017] [Indexed: 11/20/2022] Open
Abstract
Human T-cell leukemia virus type-1 (HTLV-1) causes two distinct diseases, adult T-cell leukemia/lymphoma (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Since there are no disease-specific differences among HTLV-1 strains, the etiological mechanisms separating these respective lymphoproliferative and inflammatory diseases are not well understood. In this study, by using IL-2-dependent HTLV-1-infected T-cell lines (ILTs) established from patients with ATL and HAM/TSP, we demonstrate that the anti-inflammatory cytokine IL-10 and its downstream signals potentially act as a switch for proliferation in HTLV-1-infected cells. Among six ILTs used, ILTs derived from all three ATL patients grew much faster than those from three HAM/TSP patients. Although most of the ILTs tested produced IFN-γ and IL-6, the production of IL-10 was preferentially observed in the rapid-growing ILTs. Interestingly, treatment with exogenous IL-10 markedly enhanced proliferation of the slow-growing HAM/TSP-derived ILTs. The IL-10-mediated proliferation of these ILTs was associated with phosphorylation of STAT3 and induction of survivin and IRF4, all of which are characteristics of ATL cells. Knockdown of STAT3 reduced expression of IL-10, implying a positive-feedback regulation between STAT3 and IL-10. STAT3 knockdown also reduced survivin and IRF4 in the IL-10- producing or IL-10- treated ILTs. IRF4 knockdown further suppressed survivin expression and the cell growth in these ILTs. These findings indicate that the IL-10-mediated signals promote cell proliferation in HTLV-1-infected cells through the STAT3 and IRF4 pathways. Our results imply that, although HTLV-1 infection alone may not be sufficient for cell proliferation, IL-10 and its signaling pathways within the infected cell itself and/or its surrounding microenvironment may play a critical role in pushing HTLV-1-infected cells towards proliferation at the early stages of HTLV-1 leukemogenesis. This study provides useful information for understanding of disease mechanisms and disease-prophylactic strategies in HTLV-1 infection.
Collapse
Affiliation(s)
- Leila Sawada
- Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Bunkyo-ku, Tokyo, Japan
| | - Yoshiko Nagano
- Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Bunkyo-ku, Tokyo, Japan
| | - Atsuhiko Hasegawa
- Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Bunkyo-ku, Tokyo, Japan
| | - Hikari Kanai
- Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Bunkyo-ku, Tokyo, Japan
| | - Kai Nogami
- Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Bunkyo-ku, Tokyo, Japan
| | - Sayaka Ito
- Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Bunkyo-ku, Tokyo, Japan
- Department of Medical Technology, School of Health Sciences, Tokyo University of Technology, Ota-ku, Tokyo, Japan
| | - Tomoo Sato
- Department of Rare Disease Research, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Yoshihisa Yamano
- Department of Rare Disease Research, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Yuetsu Tanaka
- Department of Immunology, Graduate school of Medicine, University of the Ryukyus, Nishihara-cho, Okinawa, Japan
| | - Takao Masuda
- Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Bunkyo-ku, Tokyo, Japan
| | - Mari Kannagi
- Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
24
|
Valverde-Villegas JM, de Medeiros RM, Almeida SEM, Chies JAB. Immunogenetic profiling of 23 SNPs of cytokine and chemokine receptor genes through a minisequencing technique: Design, development and validation. Int J Immunogenet 2017; 44:135-144. [PMID: 28374494 DOI: 10.1111/iji.12314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 02/28/2017] [Indexed: 12/15/2022]
Abstract
The minisequencing technique offers accuracy and robustness to genotyping of polymorphic DNA variants, being an excellent option for the identification and analyses of prognostic/susceptibility markers in human diseases. Two multiplex minisequencing assays were designed and standardized to screen 23 candidate SNPs in cytokine, chemokine receptor and ligand genes previously associated with susceptibility to cancer and autoimmune disorders as well as to infectious diseases outcome. The SNPs were displayed in two separate panels (panel 1-IL2 rs2069762, TNFα rs1800629, rs361525; IL4 rs2243250; IL6 rs1800795; IL10 rs1800896, rs1800872; IL17A rs8193036, rs2275913 and panel 2-CCR3 rs309125, CCR4 rs6770096, rs2228428; CCR6 rs968334; CCR8 rs2853699; CXCR3 rs34334103, rs2280964;CXCR6 rs223435, rs2234358; CCL20 rs13034664, rs6749704; CCL22 rs4359426; CXCL10/IP-10 rs3921, rs56061981). A total of 305 DNA samples from healthy individuals were genotyped by minisequencing. To validate the minisequencing technique and to encompass the majority of the potential genotypes for all 23 SNPs, 20 of these samples were genotyped by Sanger sequencing. The results of both techniques were 100% in agreement. The technique of minisequencing showed high accuracy and robustness, avoiding the need for high quantities of DNA template samples. It was easily to be conducted in bulk samples derived from a highly admixed human population, being therefore an excellent option for immunogenetic studies.
Collapse
Affiliation(s)
- J M Valverde-Villegas
- Post Graduation Program in Genetic and Molecular Biology, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - R M de Medeiros
- Post Graduation Program in Genetic and Molecular Biology, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.,Technological and Scientific Development Center - CDCT, State Foundation in Production and Health Research - FEPPS, Porto Alegre, RS, Brazil
| | - S E M Almeida
- Post Graduation Program in Genetic and Molecular Biology, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.,Technological and Scientific Development Center - CDCT, State Foundation in Production and Health Research - FEPPS, Porto Alegre, RS, Brazil
| | - J A B Chies
- Post Graduation Program in Genetic and Molecular Biology, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
25
|
Mijac D, Petrovic IV, Djuranovic S, Perovic V, Bojic D, Culafic D, Popovic D, Krstic M, Jankovic G, Djoric M, Pravica V, Markovic M. The Polymorphism rs3024505 (C/T) Downstream of the IL10 Gene Is Associated with Crohn's Disease in Serbian Patients with Inflammatory Bowel Disease. TOHOKU J EXP MED 2017; 240:15-24. [PMID: 27558476 DOI: 10.1620/tjem.240.15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Inflammatory bowel disease (IBD), manifesting as Crohn's disease (CD) and ulcerative colitis (UC), is characterized by recurring episodes of inflammation in gastrointestinal tract, in which aberrant production of regulatory cytokine interleukin-10 (IL-10) presumably plays important role. Single nucleotide polymorphisms (SNPs) that affect IL-10 production, such as rs1800896 (G/A) at position -1082 and rs1800871 (C/T) at position -819 in the promoter region of the IL10 gene, have been associated with CD and/or UC, but the results were inconsistent. Another SNP that may alter IL-10 production, rs3024505 (C/T) located immediately downstream of the IL10 gene has been recently identified. T allele of rs3024505 was associated with both UC and CD in Western populations, but the studies from East European countries are lacking. Therefore, our aim was to assess the association of rs3024505, rs1800896 and rs1800871 with Serbian IBD patients. To this end, 107 CD and 99 UC patients and 255 healthy controls were genotyped. As a result, T allele of rs3024505 was associated with CD at allelic, genotypic (GT genotype) and haplotypic (GCCT haplotype) level, suggesting potential role of this variant in susceptibility to CD. In contrast, CD patients carrying C allele of rs3024505 had significantly increased risk of anemia and stricturing/penetrating behavior. No association was observed between rs3024505 and UC or SNPs in IL10 promoter region and any form of IBD. In conclusion, rs3024505 SNP flanking the IL10 gene is associated with susceptibility and severity of disease in Serbian CD patients, further validating its role as a potential biomarker in IBD.
Collapse
Affiliation(s)
- Dragana Mijac
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, School of Medicine, University of Belgrade
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Jiang C, Liu S, Liu S, Li Z, Chen P, Chen L. Association Between the Interleukin-10-1082G/A, -592C/A, -819C/T Gene Polymorphism and HIV-1 Susceptibility: A Meta-Analysis. AIDS Res Hum Retroviruses 2017; 33:61-67. [PMID: 27785918 DOI: 10.1089/aid.2016.0056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The Interleukin-10 (IL-10) gene polymorphism influences the pathogenesis and evolution of HIV-1 disease. Many studies in this regard have evaluated the association between this polymorphism and HIV-1 susceptibility, yet, the exact relationship between them remains ambiguous and contradictory. A systematic literature search was conducted and the found case-control studies assessing the association between IL-10-1082G/A, -592C/A, -819C/T gene polymorphism and HIV-1 susceptibility were analyzed. The pooled odds ratios (ORs) and 95% confidence interval (CI) were calculated by a fixed effect model. In general, no significant relationship was found between IL-10-1082G/A gene polymorphism and susceptibility to HIV-1 infection (A vs. G genotype model: OR = 0.97, 95% CI = 0.81-1.23, p = .775; GG vs. AA+AG model: OR = 0.98, 95% CI = 0.76-1.27, p = .867; GG+AG vs. AA model: OR = 0.97, 95% CI = 0.70-1.35, p = .852; GG vs. AA model: OR = 0.88, 95% CI = 0.67-1.15, p = .348; AG vs. AA model: OR = 0.96, 95% CI = 0.67-1.37, p = .811; GG+AA vs. AG model: OR = 1.03, 95% CI = 0.74-1.43, p = .886). IL-10-529C/A gene polymorphism might lead to a decreased risk of HIV-1 infection (A vs. G genotype model: OR = 0.88, 95% CI = 0.73-1.06, p = .166; GG vs. AA+AG model: OR = 0.94, 95% CI = 0.80-1.11, p = .447; GG+AG vs. AA model: OR = 0.75, 95% CI = 0.61-0.92, p = .005; GG vs. AA model: OR = 0.73, 95% CI = 0.57-0.93, p = .012; AG vs. AA model: OR = 0.74, 95% CI = 0.60-0.92, p = .0.007; GG+AA vs. AG model: OR = 1.11, 95% CI = 0.72-1.71, p = .641). IL-10-819C/T gene polymorphism might lead to an increased risk of HIV-1 infection (A vs. G genotype model: OR = 1.25, 95% CI = 1.04-1.50, p = .019; GG vs. AA+AG model: OR = 1.29, 95% CI = 0.81-2.01, p = .278; GG+AG vs. AA model: OR = 1.42, 95% CI = 1.05-1.93, p = .023; GG vs. AA model: OR = 1.63, 95% CI = 1.11-2.38, p = .012; AG vs. AA model: OR = 1.32, 95% CI = 0.95-1.84, p = .094; GG+AA vs. AG model: OR = 0.92, 95% CI = 0.72-1.19, p = .537). In general, the meta-analysis found no marked association between the IL-10-1082G/A gene polymorphism and HIV-1 susceptibility, IL-10-529C/A gene polymorphism might lead to a decreased risk of HIV-1 infection, and IL-10-819C/T gene polymorphism might lead to an increased risk of HIV-1 infection.
Collapse
Affiliation(s)
- Caixiao Jiang
- Hebei Center for Disease Control and Prevention, Shijiazhuang, China
- Department of Epidemiology and Health Statistics, School of Public Health, Central South University, Changsha, China
| | - Shujun Liu
- Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang, China
| | - Shuxia Liu
- Liaocheng Center for Disease Control and Prevention, Liaocheng, China
| | - Zhanzhan Li
- Department of Epidemiology and Health Statistics, School of Public Health, Central South University, Changsha, China
| | - Peng Chen
- Xiangya Medical School, Central South University, Changsha, China
| | - Lizhang Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Central South University, Changsha, China
| |
Collapse
|
27
|
A Canonical Correlation Analysis of AIDS Restriction Genes and Metabolic Pathways Identifies Purine Metabolism as a Key Cooperator. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2016; 2016:2460184. [PMID: 27462363 PMCID: PMC4947641 DOI: 10.1155/2016/2460184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023]
Abstract
Human immunodeficiency virus causes a severe disease in humans, referred to as immune deficiency syndrome. Studies on the interaction between host genetic factors and the virus have revealed dozens of genes that impact diverse processes in the AIDS disease. To resolve more genetic factors related to AIDS, a canonical correlation analysis was used to determine the correlation between AIDS restriction and metabolic pathway gene expression. The results show that HIV-1 postentry cellular viral cofactors from AIDS restriction genes are coexpressed in human transcriptome microarray datasets. Further, the purine metabolism pathway comprises novel host factors that are coexpressed with AIDS restriction genes. Using a canonical correlation analysis for expression is a reliable approach to exploring the mechanism underlying AIDS.
Collapse
|
28
|
Beltra JC, Decaluwe H. Cytokines and persistent viral infections. Cytokine 2016; 82:4-15. [DOI: 10.1016/j.cyto.2016.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 02/11/2016] [Accepted: 02/11/2016] [Indexed: 12/14/2022]
|
29
|
El Ghannam D, Fawzy IM, Azmy E, Hakim H, Eid I. Relation of interleukin-10 Promoter Polymorphisms to Adult Chronic Immune Thrombocytopenic Purpura in a Cohort of Egyptian Population. Immunol Invest 2016; 44:616-26. [PMID: 26436850 DOI: 10.3109/08820139.2015.1064948] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Adult chronic immune thrombocytopenic purpura (chronic ITP) is an autoimmune multifactorial bleeding disorder that occurs because of enhanced peripheral platelet destruction. Treatment decisions can be challenging because the goal of treatment is to prevent severe bleeding, but the risk of bleeding can be difficult to estimate for any individual patient. OBJECTIVE This case-control study was planned to investigate the relationship of interleukin (IL)-10 promoter (IL-10-1082, -819 and -592) polymorphisms with the susceptibility, severity and outcome of adult chronic ITP in a cohort of Egyptian population. SUBJECTS AND METHODS Typing of IL-10 promoter polymorphisms was done using restriction fragment length polymorphism for 62 adult patients with chronic ITP and 73 age- and sex-matched healthy controls. RESULTS No significant differences were found between ITP patients and controls regarding the frequency of IL-10 promoter genotypes, alleles or haplotypes. IL-10-592 AA genotype and ATA (IL-10-1082, -819 and -592) haplotype were associated with severe ITP (p = 0.003, 0.043, respectively). CONCLUSION Our findings suggest that the IL-10 promoter polymorphisms are unlikely to affect the development or treatment outcome of chronic adult ITP in Egyptian population, but IL-10-592 AA genotype and IL-10 (-1082, -819 and -592) ATA haplotype may be associated with disease severity. Because ITP is a complex disease, it is recommended that a multicenter study should be done with large sample size and unified typing technique.
Collapse
Affiliation(s)
- Doaa El Ghannam
- a Department of Clinical Pathology, Faculty of Medicine , Mansoura University , Mansoura , Egypt
| | - Iman M Fawzy
- b Laboratory Medicine Department , Mansoura Fever Hospital , Mansoura , Egypt
| | - Emad Azmy
- c Department of Clinical Hematology , and
| | - Hazem Hakim
- d Internal Medicine Department, Faculty of Medicine, Mansoura University , Mansoura , Egypt
| | - Islam Eid
- d Internal Medicine Department, Faculty of Medicine, Mansoura University , Mansoura , Egypt
| |
Collapse
|
30
|
Singh S, Sharma A, Arora SK. Combination of low producer AA-genotypes in IFN-γ and IL-10 genes makes a high risk genetic variant for HIV disease progression. Cytokine 2015; 77:135-44. [PMID: 26579633 DOI: 10.1016/j.cyto.2015.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 11/03/2015] [Accepted: 11/05/2015] [Indexed: 01/14/2023]
Abstract
Rate of HIV disease progression varies considerably among individuals, host genetic makeup be one of the possible reasons. We aimed to determine association of functional single nucleotide polymorphism (SNPs), (-179G/T and +874T/A) in IFN-γ and (-1082A/G, -819C/T and -592C/A) in IL-10 genes, with the rate of disease progression or susceptibility to HIV infection. Therapy naïve HIV infected individuals from North India, categorized as slow progressors or fast progressors and HIV exposed seronegative individuals were recruited for this study. Genotyping results revealed significantly higher frequencies of low producer AA genotype at +874T/A in IFN-γ gene and -592C/A position in IL-10 gene in FPs (p<0.002). Multifactor dimensional reduction (MDR) analysis revealed this to be a high risk combination for faster disease progression in HIV-1 infected individuals. Low producer AA genotype carriers at +874T/A in IFN-γ gene produced significantly low amounts of cellular IFN-γ. Low producing haplotype 'ATA' at -1082, -819 and -592 loci in IL-10 gene was significantly over-represented in FPs as compared to SPs (p<0.01) and these individuals showed poor response to therapy in terms of CD4 count gains after one year of ART, compared to high producing haplotype (GCC) carriers. Thus, a combination of genetic variations in IFN-γ and IL-10 cytokine genes in a single host associate with HIV disease progression and may help clinicians to better manage the HIV disease if known earlier.
Collapse
Affiliation(s)
- Sukhvinder Singh
- Departments of Immunopathology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Aman Sharma
- Departments of Internal Medicine, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Sunil K Arora
- Departments of Immunopathology, Postgraduate Institute of Medical Education & Research, Chandigarh, India.
| |
Collapse
|
31
|
Demirkaya N, Wit F, Schlingemann R, Verbraak F. Neuroretinal Degeneration in HIV Patients Without Opportunistic Ocular Infections in the cART Era. AIDS Patient Care STDS 2015; 29:519-32. [PMID: 26258992 DOI: 10.1089/apc.2015.0091] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Subtle structural and functional retinal abnormalities, termed 'HIV-associated Neuroretinal Disorder (HIV-NRD)', have been reported in HIV patients receiving combination antiretroviral therapy (cART), without infectious retinitis or any apparent fundus abnormalities otherwise. In this review, we provide an overview of studies investigating HIV-NRD in HIV patients without opportunistic ocular infections in the cART era, and try to elucidate underlying mechanisms and associated risk factors. Most studies focused on patients with severe immune-deficiency and demonstrated that patients with nadir CD4 counts<100 cells/μL are most at risk for neuroretinal damage, with a thinner retinal nerve fiber layer, subtle loss of color vision and/or contrast sensitivity, visual field deficits, and subnormal electrophysiological responses. In contrast, alterations in retinal vascular calibers and retinal blood flow were not associated with nadir CD4 counts, but instead with detectable viremia, suggesting a role for (chronic) inflammation in microvascular damage. Although the alterations in visual function are subtle, they can lead to difficulties in activities, such as reading or driving, thereby affecting quality of life. Since HIV has become a chronic disease, its long-term effects with respect to visual function loss become more important, as is recently emphasized by a longitudinal study, reporting that AIDS patients with HIV-NRD have higher risks of developing bilateral visual impairment and even blindness than patients without HIV-NRD. The question remains whether patients with high (>350 cells/μL) nadir CD4 counts and well-suppressed HIV infection on cART remain at risk for HIV-NRD, as this group constitutes a growing part of the aging HIV-infected population.
Collapse
Affiliation(s)
- Nazli Demirkaya
- Department of Ophthalmology, Academic Medical Center, Amsterdam, The Netherlands
| | - Ferdinand Wit
- Departments of Global Health and Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
- Department of Internal Medicine, Division of Infectious Diseases, Center for Infection and Immunity Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | - Reinier Schlingemann
- Department of Ophthalmology, Academic Medical Center, Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience (NIN), Royal Academy of Sciences (KNAW), Amsterdam, The Netherlands
| | - Franciscus Verbraak
- Department of Ophthalmology, Academic Medical Center, Amsterdam, The Netherlands
- Department of Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Esposito S, Patria MF, Spena S, Codecà C, Tagliabue C, Zampiero A, Lelii M, Montinaro V, Pelucchi C, Principi N. Impact of genetic polymorphisms on paediatric atopic dermatitis. Int J Immunopathol Pharmacol 2015; 28:286-95. [DOI: 10.1177/0394632015591997] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In order to investigate whether polymorphisms of genes encoding some factors of innate and adaptive immunity play a role in the development of, or protection against atopic dermatitis (AD) and condition its severity, we genotyped 33 candidate genes and 47 single nucleotide polymorphisms (SNPs) using Custom TaqMan Array Microfluidic Cards and an ABI 7900HT analyser (Applied Biosystems, Foster City, CA, USA). The study involved 104 children with AD (29 with mild-to-moderate and 75 with severe disease; 42 girls; mean age ± SD, 5.8 ± 3.3 years) and 119 healthy controls (49 girls; mean age, 4.8 ± 3.0 years). IL10-rs1800872T, TG and MBL2-rs500737AG were all significantly more frequent among the children with AD ( P = 0.015, P = 0.004 and P = 0.030), whereas IL10-rs1800896C and TC were more frequent in those without AD ( P = 0.028 and P = 0.032). The VEGFA-rs2146326A and CTLA4-rs3087243AG SNPs were significantly more frequent in the children with mild/moderate AD than in those with severe AD ( P = 0.048 and P = 0.036). IL10-rs1800872T and TG were significantly more frequent in the children with AD and other allergic diseases than in the controls ( P = 0.014 and P = 0.007), whereas IL10-rs1800896TC and C were more frequent in the controls than in the children with AD and other allergic diseases ( P = 0.0055 and P = 0.0034). These findings show that some of the polymorphisms involved in the immune response are also involved in some aspects of the development and course of AD and, although not conclusive, support the immunological hypothesis of the origin of the inflammatory lesions.
Collapse
Affiliation(s)
- Susanna Esposito
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Francesca Patria
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia Spena
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Claudio Codecà
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Claudia Tagliabue
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Zampiero
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mara Lelii
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Montinaro
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Claudio Pelucchi
- Department of Epidemiology, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Nicola Principi
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
33
|
IL10 Variant g.5311A Is Associated with Visceral Leishmaniasis in Indian Population. PLoS One 2015; 10:e0124559. [PMID: 25941808 PMCID: PMC4420251 DOI: 10.1371/journal.pone.0124559] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 03/16/2015] [Indexed: 12/20/2022] Open
Abstract
Background Visceral leishmaniasis (VL) is a multifactorial disease, where the host genetics play a significant role in determining the disease outcome. The immunological role of anti-inflammatory cytokine, Interleukin 10 (IL10), has been well-documented in parasite infections and considered as a key regulatory cytokine for VL. Although VL patients in India display high level of IL10 in blood serum, no genetic study has been conducted to assess the VL susceptibility / resistance. Therefore, the aim of this study is to investigate the role of IL10 variations in Indian VL; and to estimate the distribution of disease associated allele in diverse Indian populations. Methodology All the exons and exon-intron boundaries of IL10 were sequenced in 184 VL patients along with 172 ethnically matched controls from VL endemic region of India. Result and Discussion Our analysis revealed four variations; rs1518111 (2195 A>G, intron), rs1554286 (2607 C>T, intron), rs3024496 (4976 T>C, 3’ UTR) and rs3024498 (5311 A>G, 3’ UTR). Of these, a variant g.5311A is significantly associated with VL (χ2=18.87; p =0.00001). In silico approaches have shown that a putative micro RNA binding site (miR-4321) is lost in rs3024498 mRNA. Further, analysis of the above four variations in 1138 individuals from 34 ethnic populations, representing different social and linguistic groups who are inhabited in different geographical regions of India, showed variable frequency. Interestingly, we have found, majority of the tribal populations have low frequency of VL (‘A’ of rs3024498); and high frequency of leprosy (‘T’ of rs1554286), and Behcet’s (‘A’ of rs1518111) associated alleles, whereas these were vice versa in castes. Our findings suggest that majority of tribal populations of India carry the protected / less severe allele against VL, while risk / more severe allele for leprosy and Behcet’s disease. This study has potential implications in counseling and management of VL and other infectious diseases.
Collapse
|
34
|
da Silva NMO, Germano FN, Vidales-Braz BM, Carmo Zanella RD, dos Santos DM, Lobato R, de Martinez AMB. Polymorphisms of IL-10 gene in patients infected with HCV under antiviral treatment in southern Brazil. Cytokine 2015; 73:253-7. [PMID: 25797191 DOI: 10.1016/j.cyto.2014.12.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/23/2014] [Accepted: 12/27/2014] [Indexed: 12/20/2022]
Abstract
Interleukin-10 (IL-10) is a cytokine that plays an important role in the regulation of the immune system. Gene polymorphisms of IL-10 have been associated with the different expression levels of this cytokine. In hepatitis C virus infection, IL-10 appears to interfere with the progression of disease, viral persistence and the response to therapy. This study investigated genetic variability in the IL-10 gene promoter between patients infected with hepatitis C virus (HCV) and healthy individuals, associating the frequency of polymorphisms with different aspects of viral infection. This is a case-control study with 260 patients who were infected with HCV and 260 healthy individuals. Genotyping of the polymorphisms was performed using the technique of amplification refractory mutation system PCR (ARMS-PCR) for regions of the IL-10 gene promoter (-1082 G/A, -819 C/T, -592 C/A). The frequencies of alleles and genotypes related to polymorphisms in the IL-10 gene promoter showed a higher frequency of the G allele and genotype GG in the -1082 region between the infected group and the control group (p=0.005 and p=0.001, respectively), whereas the AA genotype was significantly more frequent in the control group. The frequencies of the haplotypes GTA and GCC were higher in the group of infected individuals, whereas the haplotype ATA was more frequent in the healthy group (p<0.006). It was also observed that the genotypes GG and AG in the region -1082 were significantly more frequent among patients infected with HCV who were in advanced stages of fibrosis and cirrhosis (p=0.042). No association was observed between polymorphisms of IL-10 and sustained virologic response (SVR).
Collapse
|
35
|
Polymorphisms in the IFNγ, IL-10, and TGFβ genes may be associated with HIV-1 infection. DISEASE MARKERS 2015; 2015:248571. [PMID: 25802474 PMCID: PMC4354727 DOI: 10.1155/2015/248571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 01/11/2023]
Abstract
OBJECTIVE This study investigated possible associations between the TNFα-308G/A, IFN+874A/T, IL-6-174C/G, IL-10-1082A/G, and TGFβ-509C/T polymorphisms with HIV-1 infection, in addition to correlation of the polymorphisms with clinical markers of AIDS progression, such as levels of CD4+/CD8+ T lymphocytes and plasma viral load. METHODS A total of 216 individuals who were infected with HIV-1 and on antiretroviral therapy (ART) and 294 individuals from the uninfected control group were analyzed. RESULTS All individuals evaluated were negative for total anti-HBc, anti-HCV, anti-T. pallidum, and anti-HTLV-1/2. The polymorphisms were identified by PCR-RFLP. Individuals presenting the IFN+874A allele as well as the AA genotype were more frequent in the HIV-1 infected group compared to the control group (P < 0.05), in addition to having lower levels of CD4+ T lymphocytes. The CD8+ T lymphocytes count was significantly lower in individuals with the IL-10-1082 GG genotype. The TGFβ-509TT genotype was associated with higher plasma viral load. CONCLUSIONS The results suggest that the presence of the IFN+874A allele confers susceptibility to HIV-1 infection and a decrease in the number of CD4+ T lymphocytes. In addition, the genotype associated with high serum levels of TGFβ may be associated with an increase in plasma viral load.
Collapse
|
36
|
Immunogenetic influences on acquisition of HIV-1 infection: consensus findings from two African cohorts point to an enhancer element in IL19 (1q32.2). Genes Immun 2015; 16:213-20. [PMID: 25633979 PMCID: PMC4409473 DOI: 10.1038/gene.2014.84] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/19/2014] [Accepted: 12/19/2014] [Indexed: 12/12/2022]
Abstract
Numerous reports have suggested that immunogenetic factors may influence HIV-1 acquisition, yet replicated findings that translate between study cohorts remain elusive. Our work aimed to test several hypotheses about genetic variants within the IL10-IL24 gene cluster that encodes interleukin (IL)-10, IL-19, IL-20, and IL-24. In aggregated data from 515 Rwandans and 762 Zambians with up to 12 years of follow-up, 190 single nucleotide polymorphisms (SNPs) passed quality control procedures. When HIV-1-exposed seronegative subjects (n = 486) were compared with newly seroconverted individuals (n = 313) and seroprevalent subjects (n = 478) who were already infected at enrollment, rs12407485 (G>A) in IL19 showed a robust association signal in adjusted logistic regression models (odds ratio = 0.64, P = 1.7 × 10−4, and q = 0.033). Sensitivity analyses demonstrated that (i) results from both cohorts and subgroups within each cohort were highly consistent; (ii) verification of HIV-1 infection status after enrollment was critical; and (iii) supporting evidence was readily obtained from Cox proportional hazards models. Data from public databases indicate that rs12407485 is part of an enhancer element for three transcription factors. Overall, these findings suggest that molecular features at the IL19 locus may modestly alter the establishment of HIV-1 infection.
Collapse
|
37
|
Evaluation of Interleukin-10 Levels in Patients Diagnosed with Chronic Hepatitis. W INDIAN MED J 2015; 64:71-5. [PMID: 26360676 DOI: 10.7727/wimj.2014.154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 06/30/2014] [Indexed: 01/12/2023]
Abstract
OBJECTIVE One of the most important factors playing a role in chronic hepatitis B pathogenesis is cytokine release and one of the cytokines with anti-inflammatory characteristic is interleukin-10 (IL-10). The aim of the present study is to examine IL-10 levels in patients with chronic hepatitis B. SUBJECTS AND METHODS Sixty-three patients with chronic hepatitis B disease who had not received any antiviral treatment were included in the study. Serum IL-10 level was investigated by enzyme-linked immunosorbent assay (ELISA) method. In the control group, 25 healthy individuals with mean age similar to the patient population were included. Control and patient groups were compared and data were statistically analysed. RESULTS Interleukin-10 levels of 25 patients with hepatitis B virus (HBV) DNA levels between 2000 and 20 000 IU/mL were compared with those of 25 subjects in the control group, and the level in the chronic hepatitis B group was statistically significantly higher (p < 0.05). Interleukin-10 levels of 38 patients with HBV DNA > 20 000 IU/mL were statistically significantly higher than those in the control group. When chronic hepatitis B patients were compared among themselves, IL-10 levels increased as HBV DNA levels increased. Also, when IL-10 levels of hepatitis B 'e' antigen (HBeAg) positive patients were compared with those of HBeAg negative patients, the difference was not statistically significant. CONCLUSION It is believed that decreasing IL-10 levels by various methods would have significant contributions in disease progression and treatment. Moreover, IL-10 level may be an important marker in HBeAg seroconversion and evaluation of treatment response.
Collapse
|
38
|
Ramezani A, Kalantar E, Aghakhani A, Banifazl M, Foroughi M, Hosseini S, Eslamifar A, Esmaeilzadeh A, Mohraz M. Lack of Association between Interleukin-10 Gene Promoter Polymorphisms with HIV Susceptibility and Progression to AIDS. IRANIAN JOURNAL OF PATHOLOGY 2015; 10:141-148. [PMID: 26351475 PMCID: PMC4539764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 08/02/2014] [Indexed: 06/05/2023]
Abstract
BACKGROUND Interleukin (IL)-10 is an important anti-inflammatory and immunomodulatory cytokine. Some authors believe that single nucleotide polymorphisms (SNP) in the promoter region of the IL-10 gene have been associated with susceptibility to HIV infection and progression to AIDS, but its role is not clearly defined yet. The present study was undertaken to evaluate the association between HIV infection susceptibility and progression with SNP in the promoter region of the IL-10 gene. METHODS This study was carried out in 70 HIV infected patients (39 treatment naïve and 31 under treatment) and 31 matched healthy controls. The biallelic polymorphisms in the IL-10 gene promoter (-592 ,-1082) were analyzed by polymerase chain reaction and direct sequencing. RESULTS At position -1082, G/A was the most common genotype and A was the most prevalent allele and at position -592, A/C was the most prevalent genotype and -592 C was the most common allele in HIV positive patients; although there was not any significant difference between cases and controls regarding genotypes and alleles of these regions. CONCLUSION Our study showed that genetic polymorphisms of IL-10 promoter region may not associate with HIV infection outcome and the lack of this association suggests that other genes may influence on HIV infection course.
Collapse
Affiliation(s)
- Amitis Ramezani
- Dept of Clinical Research, Pasteur Institute of Iran, Tehran, Iran
- Pediatric Infectious Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Arezoo Aghakhani
- Dept of Clinical Research, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Banifazl
- Iranian Society for Support Patients With Infectious Disease, Tehran, Iran
| | | | | | - Ali Eslamifar
- Dept of Clinical Research, Pasteur Institute of Iran, Tehran, Iran
| | | | - Minoo Mohraz
- Iranian Research Center for HIV/AIDS, Tehran, Iran
| |
Collapse
|
39
|
Kallas E, Huik K, Pauskar M, Jõgeda EL, Karki T, Des Jarlais D, Uusküla A, Avi R, Lutsar I. Influence of interleukin 10 polymorphisms -592 and -1082 to the HIV, HBV and HCV serostatus among intravenous drug users. INFECTION GENETICS AND EVOLUTION 2014; 30:175-180. [PMID: 25542814 DOI: 10.1016/j.meegid.2014.12.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 12/10/2014] [Accepted: 12/18/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND Interleukin 10 (IL-10) is a multifunctional cytokine produced by macrophages, monocytes, and T-helper cells. Two polymorphisms at positions -592 and -1082 have been associated with HIV susceptibility. However, their associations with susceptibility to HIV and its co-infections among intravenous drug users (IDUs) are largely unknown. METHODS A total of 345 IDUs were recruited. Of the 173 HIV negative IDUs, 20 were classified as highly exposed HIV seronegative subjects (HESNs). A control group consisted of 496 blood donors; all HIV, HCV, and HBV negative. The IL-10-592C/A and -1082A/G were determined using TaqMan allelic discrimination assay. RESULTS Of the IDUs, 50% were HIV positive, 89% HCV positive, 67% HBV positive and 41% had triple infection. IL-10-592C allele and -1082A allele were the most common and the -1082AG/-592CC was the most common genotype pair. All HESNs exhibited -1082A allele as compared to 81.4% of the HIV positive IDUs and 79% of donors (p=0.029 and p=0.019, respectively). None of HESNs had GG/CC genotype pair compared with 18.6% of HIV positive IDUs and 21.0% of donors (p=0.029 and p=0.019, respectively). The possession of -592AC and genotype pair AG/AC were associated with the decreased odds of HBV infection (OR=0.28; 95% CI 0.09-0.87; p=0.028 and OR=0.19; 95% CI 0.06-0.61; p=0.052, respectively). CONCLUSIONS The presence of low producing IL-10-1082A and -592A alleles and their containing genetic variants protect highly exposed IDUs against acquisition of HIV and HBV infections.
Collapse
Affiliation(s)
- Eveli Kallas
- Department of Microbiology, Faculty of Medicine, University of Tartu, Tartu, Estonia(1).
| | - Kristi Huik
- Department of Microbiology, Faculty of Medicine, University of Tartu, Tartu, Estonia(1)
| | - Merit Pauskar
- Department of Microbiology, Faculty of Medicine, University of Tartu, Tartu, Estonia(1)
| | - Ene-Ly Jõgeda
- Department of Microbiology, Faculty of Medicine, University of Tartu, Tartu, Estonia(1)
| | - Tõnis Karki
- Department of Microbiology, Faculty of Medicine, University of Tartu, Tartu, Estonia(1)
| | | | - Anneli Uusküla
- Department of Public Health, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Radko Avi
- Department of Microbiology, Faculty of Medicine, University of Tartu, Tartu, Estonia(1)
| | - Irja Lutsar
- Department of Microbiology, Faculty of Medicine, University of Tartu, Tartu, Estonia(1)
| |
Collapse
|
40
|
Rahimi H, Farajollahi MM, Hosseini A. Distribution of the mutated delta 32 allele of CCR5 co-receptor gene in Iranian population. Med J Islam Repub Iran 2014; 28:140. [PMID: 25694998 PMCID: PMC4322332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/14/2014] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The CCR5 is a chemokine receptor that serves as a co-receptor for HIV-1 attachment and entry to T lymphocytes. A 32bp deletion (∆32) in this gene is believed to be associated with resistance to infection and delay disease progression. The aim of this study was to determine the∆32 allele frequency in healthy individuals and HIV-infected individuals with AIDS. METHODS In this experiment, 530 normal individuals from healthy Iranian population and 40 HIV-infected samples from Western Clinic of Tehran were examined for∆32 in CCR5 gene using polymerase chain reaction (PCR) techniques followed by agarose gel electrophoresis. RESULTS Allele frequencies of the CCR5∆32 in normal individuals were calculated to be 1.1% for heterozygous genotype and 0.19% for homozygous genotype. None of the co-receptor gene in HIV cases was found to be mutated in this study. CONCLUSION Based on the findings of this study and the literature in Iran, we could conclude that Iranian people similar to neighbor countries such as Arabs are susceptible to HIV virus infection.
Collapse
Affiliation(s)
- Hamzeh Rahimi
- 1. PhD student of Department of Medical Biotechnology, Iranian Pasteur Institute, Pasteur Square, Tehran, Iran.
| | - Mohammad M Farajollahi
- 2. Professor of Department of Medical Biotechnology, Faculty of Allied Medicine & Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran & Alborz Food & Drug Laboratory, Fardis, Karaj, Alborz, Iran.
| | - Arshad Hosseini
- 3. Assistant Professor of Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
41
|
Jaskula E, Lange A, Dlubek D, Kyrcz-Krzemień S, Markiewicz M, Dzierzak-Mietla M, Jedrzejczak WW, Gronkowska A, Nowak J, Warzocha K, Hellmann A, Kowalczyk J, Drabko K, Goździk J, Mizia S. IL-10 promoter polymorphisms influence susceptibility to aGvHD and are associated with proportions of CD4+FoxP3+ lymphocytes in blood after hematopoietic stem cell transplantation. ACTA ACUST UNITED AC 2014; 82:387-96. [PMID: 24498995 DOI: 10.1111/tan.12255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 09/13/2013] [Accepted: 10/21/2013] [Indexed: 01/17/2023]
Abstract
Four hundred and ninety-five patients (390 and 105 grafted from unrelated and sibling (SIB) donors, respectively) and their donors were analyzed for the impact of interleukin-10 (IL-10) promoter genotype [rs18000896 (-1082 G/A), rs18000871 (-819 C/T) and rs18000872 (-592 C/A)] on the outcome of hematopoietic stem cell transplantation (HSCT). Patients having ACC haplotype were at a lower risk of acute graft versus host disease (aGvHD, grade > I) if transplanted from human leukocyte antigen (HLA) well-matched (10/10) unrelated donors (20/135 vs 39/117, P < 0.001, Pcorr = 0.002), which was not seen if patients were transplanted from either sibling (SIB) or poorly matched (<10/10) unrelated donors (MUD). In addition, GCC haplotype positive recipients of unrelated donor transplants tended to be more susceptible to aGvHD (68/199 vs 39/169, P = 0.019, Pcorr = 0.057). Multivariate logistic regression analysis in the MUD transplanted group showed that donor-recipient human leukocyte antigen (HLA) mismatch [odds ratio (OR) = 3.937, P = 0.001] and a lack of ACC haplotype in recipients (OR = 0.417, P = 0.013) played a significant role as independent risk factors of aGvHD grade > I. ACC carriers had higher proportions of FoxP3+ lymphocytes gated in CD4+ lymphocytes as compared with patients with other IL-10 haplotypes. It was seen at the time of hematological recovery (mean ± SEM: 3.80 ± 0.91% vs 2.06 ± 0.98%, P = 0.012) and 2 weeks later (5.32 ± 0.87% vs 2.50 ± 0.83%, P = 0.013); -592 C/A polymorphism was separately analyzed and it was found that AA homozygotes tended to have a higher incidence of aGvHD (8/15 vs 116/456, P = 0.034) and low proportions of FoxP3 CD4+ lymphocytes in blood (0.43 ± 0.22% vs 4.32 ± 0.71%, P = 0.051) measured 2 weeks after hematological recovery. Functional IL-10 polymorphism associated features influenced the risk of aGvHD with a positive effect of ACC on the pool of Treg in blood.
Collapse
Affiliation(s)
- E Jaskula
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Cheng SB, Sharma S. Interleukin-10: a pleiotropic regulator in pregnancy. Am J Reprod Immunol 2014; 73:487-500. [PMID: 25269386 DOI: 10.1111/aji.12329] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/09/2014] [Indexed: 12/14/2022] Open
Abstract
Pregnancy is a unique and well-choreographed physiological process that involves intricate interplay of inflammatory and anti-inflammatory milieu, hormonal changes, and cellular and molecular events at the maternal-fetal interface. IL-10 is a pregnancy compatible cytokine that plays a vital role in maintaining immune tolerance. A wide array of cell types including both immune and non-immune cells secret IL-10 in an autocrine and paracrine manner. IL-10 binds to a specific receptor complex and activates JAK-STAT and PI3K-Akt signaling pathways while inhibiting NF-κB signaling pathway. IL-10 exerts its anti-inflammatory effects mainly by decreasing pro-inflammatory cytokines such as IL-1, IL-6, IL-12, and TNF-α, by inducing heme oxygenase-1, and by inhibiting antigen presentation via blocking major histocompatibility complex (MHC) class II expression. Prior studies from our group and others have shown that IL-10 also functions as a potent protector against vascular dysfunction, and enhancement of IL-10 may serve as an immunotherapeutic intervention to treat adverse pregnancy outcomes. This review seeks to critically evaluate the archetypal functions of IL-10 as an immune suppressive factor as well as its novel functions as a vascular protector and modulator of endoplasmic reticulum (ER) stress and autophagy in the context of normal and adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Shi-Bin Cheng
- Department of Pediatrics, Women and Infants' Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Surendra Sharma
- Department of Pediatrics, Women and Infants' Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
43
|
Corchado S, López-Cortés LF, Rivero-Juárez A, Torres-Cornejo A, Rivero A, Márquez-Coello M, Girón-González JA. Liver fibrosis, host genetic and hepatitis C virus related parameters as predictive factors of response to therapy against hepatitis C virus in HIV/HCV coinfected patients. PLoS One 2014; 9:e101760. [PMID: 25013899 PMCID: PMC4094489 DOI: 10.1371/journal.pone.0101760] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 06/11/2014] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE To establish the role of liver fibrosis as a predictive tool of response to pegylated interferon alpha (Peg-IFN) and ribavirin (RBV) treatment in human immunodeficiency (HIV)/hepatitis C virus (HCV) coinfected patients, in addition to recognized predictive factors (HCV load, HCV genotype, IL-28B polymorphism). PATIENTS AND METHODS A sample of 267 HIV/HCV coinfected patients was treated with Peg-IFN and RBV. Predictive factors of rapid (RVR) and sustained (SVR) virological response were analyzed. Independent variables were age, sex, IL28B, -238 TNF-α and -592 IL-10 polymorphisms, HCV genotype, HCV-RNA levels, significant fibrosis or cirrhosis and CD4+ T cell count. RESULTS Patients infected by HCV genotype 1 (n = 187) showed RVR and SVR in 12% and 39% of cases, respectively. The parameters associated with RVR were IL28B genotype CC and plasma HCV-RNA levels <600,000 IU/ml. Advanced liver fibrosis was negatively associated with SVR in patients without RVR. A SVR was obtained in 42% of subjects with HCV genotype 4, and the independent factors associated with SVR were IL28B genotype CC and an HCV-RNA <600,000 IU/ml. A SVR was obtained in 66% of patients with HCV genotypes 2/3; in this case, the independent parameter associated with SVR was the absence of significant liver fibrosis. TNF-α and IL-10 polymorphisms were not associated with SVR, although a significantly higher percentage of -238 TNF-α genotype GG was detected in patients with significant liver fibrosis. CONCLUSIONS In HIV/HCV coinfected patients with HCV genotypes 1 or 4, RVR, mainly influenced by genotype IL28B and HCV-RNA levels, reliably predicted SVR after 4 weeks of therapy with Peg-IFN plus RBV. In patients infected by HCV genotype 3, an elevated relapse rate compromised the influence of RVR on SVR. Relapses were related to the presence of advanced liver fibrosis. Liver cirrhosis was associated with a -238 TNF-α polymorphism in these patients.
Collapse
Affiliation(s)
- Sara Corchado
- Unidad de Enfermedades Infecciosas, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Luis F. López-Cortés
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/Centro Superior de Investigaciones Científicas/Universidad de Sevilla, Sevilla, Spain
| | - Antonio Rivero-Juárez
- Maimonides Institute for Research in Biomedicine of Cordoba/Reina Sofia University Hospital, Córdoba, Spain
| | - Almudena Torres-Cornejo
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/Centro Superior de Investigaciones Científicas/Universidad de Sevilla, Sevilla, Spain
| | - Antonio Rivero
- Maimonides Institute for Research in Biomedicine of Cordoba/Reina Sofia University Hospital, Córdoba, Spain
| | | | | |
Collapse
|
44
|
Saxena R, Chawla YK, Verma I, Kaur J. Association of interleukin-10 with hepatitis B virus (HBV) mediated disease progression in Indian population. Indian J Med Res 2014; 139:737-45. [PMID: 25027084 PMCID: PMC4140039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND & OBJECTIVES Interleukin (IL)-10, an anti-inflammatory Th2 cytokine, is one of the key coordinators of the inflammatory responses involved. The present study was designed to evaluate the impact of IL-10 (-819/-592) genotypes, haplotypes, mRNA and the protein levels with risk for hepatitis B virus (HBV) related hepatocellular carcinoma (HCC) development in India. METHODS A total of 390 subjects (145 controls, 62 inactive HBV-carriers, 64 chronic-active HBV patients, 60 HBV related cirrhotics and 59 HBV- HCC subjects) were enrolled in the study. Allele specific (AS)-PCR, ELISA and RT-PCR methods were used for assessing polymorphism, spontaneous blood levels and the mRNA expression, respectively of IL-10. RESULTS The study revealed that the CC/TA genotype acted as a risk factor for cirrhosis (OR a =2.02; P<0.05) and the subsequent HCC development (OR a =2.20; P<0.05), with controls as reference. However, no significant association was found between the two haplotypes (CC and TA) observed and HCC risk. Moreover, the IL-10 protein and mRNA levels in peripheral blood mono nuclear cells (PBMCs) showed a significant elevation as the disease progressed to cirrhosis. But, no variation was observed in the IL-10 levels in subjects with different IL-10 genotypes. INTERPRETATION & CONCLUSIONS These preliminary results suggest a strong association of IL10 (-819/-592) with the HBV infection mediated disease progression, from inactive carrier state to malignancy, in Indian population.
Collapse
Affiliation(s)
- Roli Saxena
- Department of Biochemistry, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Yogesh Kumar Chawla
- Department of Hepatology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Indu Verma
- Department of Biochemistry, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Jyotdeep Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education & Research, Chandigarh, India,Reprint requests: Dr Jyotdeep Kaur, Department of Biochemistry, Postgraduate Institute of Medical Education & Research, Chandigarh 160 012, India e-mail:
| |
Collapse
|
45
|
Hauser KF, Knapp PE. Interactions of HIV and drugs of abuse: the importance of glia, neural progenitors, and host genetic factors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 118:231-313. [PMID: 25175867 PMCID: PMC4304845 DOI: 10.1016/b978-0-12-801284-0.00009-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Considerable insight has been gained into the comorbid, interactive effects of HIV and drug abuse in the brain using experimental models. This review, which considers opiates, methamphetamine, and cocaine, emphasizes the importance of host genetics and glial plasticity in driving the pathogenic neuron remodeling underlying neuro-acquired immunodeficiency syndrome and drug abuse comorbidity. Clinical findings are less concordant than experimental work, and the response of individuals to HIV and to drug abuse can vary tremendously. Host-genetic variability is important in determining viral tropism, neuropathogenesis, drug responses, and addictive behavior. However, genetic differences alone cannot account for individual variability in the brain "connectome." Environment and experience are critical determinants in the evolution of synaptic circuitry throughout life. Neurons and glia both exercise control over determinants of synaptic plasticity that are disrupted by HIV and drug abuse. Perivascular macrophages, microglia, and to a lesser extent astroglia can harbor the infection. Uninfected bystanders, especially astroglia, propagate and amplify inflammatory signals. Drug abuse by itself derails neuronal and glial function, and the outcome of chronic exposure is maladaptive plasticity. The negative consequences of coexposure to HIV and drug abuse are determined by numerous factors including genetics, sex, age, and multidrug exposure. Glia and some neurons are generated throughout life, and their progenitors appear to be targets of HIV and opiates/psychostimulants. The chronic nature of HIV and drug abuse appears to result in sustained alterations in the maturation and fate of neural progenitors, which may affect the balance of glial populations within multiple brain regions.
Collapse
Affiliation(s)
- Kurt F Hauser
- Department of Pharmacology & Toxicology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA.
| | - Pamela E Knapp
- Department of Pharmacology & Toxicology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA; Department of Anatomy & Neurobiology, Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
46
|
Damodharan S, Gujar R, Pattabiraman S, Nesakumar M, Hanna LE, Vadakkuppattu RD, Usha R. Expression and immunological characterization of cardamom mosaic virus coat protein displaying HIV gp41 epitopes. Microbiol Immunol 2013; 57:374-85. [PMID: 23668610 DOI: 10.1111/1348-0421.12045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/22/2013] [Accepted: 03/13/2013] [Indexed: 12/14/2022]
Abstract
The coat protein of cardamom mosaic virus (CdMV), a member of the genus Macluravirus, assembles into virus-like particles when expressed in an Escherichia coli expression system. The N and C-termini of the coat protein were engineered with the Kennedy peptide and the 2F5 and 4E10 epitopes of gp41 of HIV. The chimeric proteins reacted with sera from HIV positive persons and also stimulated secretion of cytokines by peripheral blood mononuclear cells from these persons. Thus, a system based on the coat protein of CdMV can be used to display HIV-1 antigens.
Collapse
Affiliation(s)
- Subha Damodharan
- Department of Plant Biotechnology, School of Biotechnology, Madurai Kamaraj University, Palkalainagar, Madurai 625021, India
| | | | | | | | | | | | | |
Collapse
|
47
|
Richter K, Perriard G, Behrendt R, Schwendener RA, Sexl V, Dunn R, Kamanaka M, Flavell RA, Roers A, Oxenius A. Macrophage and T cell produced IL-10 promotes viral chronicity. PLoS Pathog 2013; 9:e1003735. [PMID: 24244162 PMCID: PMC3820745 DOI: 10.1371/journal.ppat.1003735] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 09/13/2013] [Indexed: 01/12/2023] Open
Abstract
Chronic viral infections lead to CD8+ T cell exhaustion, characterized by impaired cytokine secretion. Presence of the immune-regulatory cytokine IL-10 promotes chronicity of Lymphocytic Choriomeningitis Virus (LCMV) Clone 13 infection, while absence of IL-10/IL-10R signaling early during infection results in viral clearance and higher percentages and numbers of antiviral, cytokine producing T cells. IL-10 is produced by several cell types during LCMV infection but it is currently unclear which cellular sources are responsible for induction of viral chronicity. Here, we demonstrate that although dendritic cells produce IL-10 and overall IL-10 mRNA levels decrease significantly in absence of CD11c+ cells, absence of IL-10 produced by CD11c+ cells failed to improve the LCMV-specific T cell response and control of LCMV infection. Similarly, NK cell specific IL-10 deficiency had no positive impact on the LCMV-specific T cell response or viral control, even though high percentages of NK cells produced IL-10 at early time points after infection. Interestingly, we found markedly improved T cell responses and clearance of normally chronic LCMV Clone 13 infection when either myeloid cells or T cells lacked IL-10 production and mice depleted of monocytes/macrophages or CD4+ T cells exhibited reduced overall levels of IL-10 mRNA. These data suggest that the decision whether LCMV infection becomes chronic or can be cleared critically depends on early CD4+ T cell and monocyte/macrophage produced IL-10. Chronic viral infections like Hepatitis B and C Virus (HBV and HCV) and Human Immunodeficiency Virus (HIV) in humans affect more than 500 million people worldwide. While a robust T cell response is a hallmark of many acute infections one hurdle inhibiting the clearance of chronic viral infections is that the immune-suppressive cytokine IL-10 modulates the virus-host balance towards induction of T cell dysfunction. IL-10 is produced by several cell types during chronic Lymphocytic Choriomeningitis Virus (LCMV) infection but it is currently unclear which cellular sources are responsible to promote viral chronicity. Here, we demonstrate that T cell responses improved markedly, and that normally chronic LCMV Clone 13 infection could be cleared when either myeloid cells or T cells lacked IL-10 production. Furthermore, mice depleted of monocytes/macrophages or CD4+ T cells exhibited reduced overall levels of IL-10 mRNA. These data suggest that the decision whether LCMV infection becomes chronic or can be cleared critically depends on CD4+ T cell and monocyte/macrophage produced IL-10 early during the establishment of viral chronicity.
Collapse
Affiliation(s)
| | | | - Rayk Behrendt
- Institute of Immunology, Technical University of Dresden, Dresden, Germany
| | - Reto A. Schwendener
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna,Vienna, Austria
| | - Robert Dunn
- Biogen Idec, San Diego, California, United States of America
| | - Masahito Kamanaka
- Department of Immunobiology and Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Richard A. Flavell
- Department of Immunobiology and Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Axel Roers
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Annette Oxenius
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
48
|
Ballana E, Esté JA. Insights from host genomics into HIV infection and disease: Identification of host targets for drug development. Antiviral Res 2013; 100:473-86. [PMID: 24084487 DOI: 10.1016/j.antiviral.2013.09.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/17/2013] [Accepted: 09/20/2013] [Indexed: 01/11/2023]
Abstract
HIV susceptibility and disease progression show a substantial degree of individual heterogeneity, ranging from fast progressors to long-term non progressors or elite controllers, that is, subjects that control infection in the absence of therapy. Recent years have seen a significant increase in understanding of the host genetic determinants of susceptibility to HIV infection and disease progression, driven in large part by candidate gene studies, genome-wide association studies, genome-wide transcriptome analyses, and large-scale functional screens. These studies have identified common variants in host loci that clearly influence disease progression, characterized the scale and dynamics of gene and protein expression changes in response to infection, and provided the first comprehensive catalogue of genes and pathways involved in viral replication. This review highlights the potential of host genomic influences in antiviral therapy by pointing to promising novel drug targets but also providing the basis of the identification and validation of host mechanisms that might be susceptible targets for novel antiviral therapies.
Collapse
Affiliation(s)
- Ester Ballana
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain.
| | | |
Collapse
|
49
|
McLaren PJ, Coulonges C, Ripke S, van den Berg L, Buchbinder S, Carrington M, Cossarizza A, Dalmau J, Deeks SG, Delaneau O, De Luca A, Goedert JJ, Haas D, Herbeck JT, Kathiresan S, Kirk GD, Lambotte O, Luo M, Mallal S, van Manen D, Martinez-Picado J, Meyer L, Miro JM, Mullins JI, Obel N, O'Brien SJ, Pereyra F, Plummer FA, Poli G, Qi Y, Rucart P, Sandhu MS, Shea PR, Schuitemaker H, Theodorou I, Vannberg F, Veldink J, Walker BD, Weintrob A, Winkler CA, Wolinsky S, Telenti A, Goldstein DB, de Bakker PIW, Zagury JF, Fellay J. Association study of common genetic variants and HIV-1 acquisition in 6,300 infected cases and 7,200 controls. PLoS Pathog 2013; 9:e1003515. [PMID: 23935489 PMCID: PMC3723635 DOI: 10.1371/journal.ppat.1003515] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 06/07/2013] [Indexed: 11/18/2022] Open
Abstract
Multiple genome-wide association studies (GWAS) have been performed in HIV-1 infected individuals, identifying common genetic influences on viral control and disease course. Similarly, common genetic correlates of acquisition of HIV-1 after exposure have been interrogated using GWAS, although in generally small samples. Under the auspices of the International Collaboration for the Genomics of HIV, we have combined the genome-wide single nucleotide polymorphism (SNP) data collected by 25 cohorts, studies, or institutions on HIV-1 infected individuals and compared them to carefully matched population-level data sets (a list of all collaborators appears in Note S1 in Text S1). After imputation using the 1,000 Genomes Project reference panel, we tested approximately 8 million common DNA variants (SNPs and indels) for association with HIV-1 acquisition in 6,334 infected patients and 7,247 population samples of European ancestry. Initial association testing identified the SNP rs4418214, the C allele of which is known to tag the HLA-B*57:01 and B*27:05 alleles, as genome-wide significant (p = 3.6×10−11). However, restricting analysis to individuals with a known date of seroconversion suggested that this association was due to the frailty bias in studies of lethal diseases. Further analyses including testing recessive genetic models, testing for bulk effects of non-genome-wide significant variants, stratifying by sexual or parenteral transmission risk and testing previously reported associations showed no evidence for genetic influence on HIV-1 acquisition (with the exception of CCR5Δ32 homozygosity). Thus, these data suggest that genetic influences on HIV acquisition are either rare or have smaller effects than can be detected by this sample size. Comparing the frequency differences between common DNA variants in disease-affected cases and in unaffected controls has been successful in uncovering the genetic component of multiple diseases. This approach is most effective when large samples of cases and controls are available. Here we combine information from multiple studies of HIV infected patients, including more than 6,300 HIV+ individuals, with data from 7,200 general population samples of European ancestry to test nearly 8 million common DNA variants for an impact on HIV acquisition. With this large sample we did not observe any single common genetic variant that significantly associated with HIV acquisition. We further tested 22 variants previously identified by smaller studies as influencing HIV acquisition. With the exception of a deletion polymorphism in the CCR5 gene (CCR5Δ32) we found no convincing evidence to support these previous associations. Taken together these data suggest that genetic influences on HIV acquisition are either rare or have smaller effects than can be detected by this sample size.
Collapse
Affiliation(s)
- Paul J. McLaren
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Cédric Coulonges
- Laboratoire Génomique, Bioinformatique, et Applications, EA4627, Chaire de Bioinformatique, Conservatoire National des Arts et Métiers, Paris, France
- ANRS Genomic Group (French Agency for Research on AIDS and Hepatitis), Paris, France
| | - Stephan Ripke
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Leonard van den Berg
- Department of Neurology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Susan Buchbinder
- Bridge HIV, San Francisco Department of Public Health, San Francisco, California, United States of America
| | - Mary Carrington
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Andrea Cossarizza
- Department of Surgery, Medicine, Dentistry and Morphological Sciences University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Judith Dalmau
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Steven G. Deeks
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Olivier Delaneau
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Andrea De Luca
- University Division of Infectious Diseases, Siena University Hospital, Siena, Italy
- Institute of Clinical infectious Diseases, Università Cattolica del Sacro Cuore, Roma, Italy
| | - James J. Goedert
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | - David Haas
- Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Joshua T. Herbeck
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Sekar Kathiresan
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Cardiovascular Research Center and Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gregory D. Kirk
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Olivier Lambotte
- INSERM U1012, Bicêtre, France
- University Paris-Sud, Bicêtre, France
- AP-HP, Department of Internal Medicine and Infectious Diseases, Bicêtre Hospital, Bicêtre, France
| | - Ma Luo
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
- National Microbiology Laboratory, Winnipeg, Manitoba, Canada
| | - Simon Mallal
- Institute for Immunology & Infectious Diseases, Murdoch University and Pathwest, Perth, Australia
| | - Daniëlle van Manen
- Department of Experimental Immunology, Sanquin Research, Landsteiner Laboratory, and Center for Infectious Diseases and Immunity Amsterdam (CINIMA) at the Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | - Javier Martinez-Picado
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Laurence Meyer
- ANRS Genomic Group (French Agency for Research on AIDS and Hepatitis), Paris, France
- Inserm, CESP U1018, University Paris-Sud, UMRS 1018, Faculté de Médecine Paris-Sud; AP-HP, Hopital Bicêtre, Epidemiology and Public Health Service, Le Kremlin Bicêtre, France
| | - José M. Miro
- Infectious Diseases Service. Hospital Clinic – IDIBAPS, University of Barcelona, Barcelona, Spain
| | - James I. Mullins
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Niels Obel
- Department of Infectious Diseases, The National University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Stephen J. O'Brien
- Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia
| | - Florencia Pereyra
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
- Division of Infectious Disease, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Francis A. Plummer
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
- National Microbiology Laboratory, Winnipeg, Manitoba, Canada
| | - Guido Poli
- Division of Immunology, Transplantation and Infectious Diseases, Vita-Salute San Raffaele University, School of Medicine & San Raffaele Scientific Institute, Milan, Italy
| | - Ying Qi
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Pierre Rucart
- Laboratoire Génomique, Bioinformatique, et Applications, EA4627, Chaire de Bioinformatique, Conservatoire National des Arts et Métiers, Paris, France
- ANRS Genomic Group (French Agency for Research on AIDS and Hepatitis), Paris, France
| | - Manj S. Sandhu
- Genetic Epidemiology Group, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
- Non-Communicable Disease Research Group, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Patrick R. Shea
- Center for Human Genome Variation, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Hanneke Schuitemaker
- Department of Experimental Immunology, Sanquin Research, Landsteiner Laboratory, and Center for Infectious Diseases and Immunity Amsterdam (CINIMA) at the Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | - Ioannis Theodorou
- ANRS Genomic Group (French Agency for Research on AIDS and Hepatitis), Paris, France
- INSERM UMRS 945, Paris, France
| | - Fredrik Vannberg
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Jan Veldink
- Department of Neurology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bruce D. Walker
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Amy Weintrob
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Cheryl A. Winkler
- Basic Research Laboratory, Molecular Genetic Epidemiology Section, Center for Cancer Research, NCI, SAIC-Frederick, Inc., Frederick National Laboratory, Frederick, Maryland, United States of America
| | - Steven Wolinsky
- Division of Infectious Diseases, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Amalio Telenti
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
| | - David B. Goldstein
- Center for Human Genome Variation, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Paul I. W. de Bakker
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Division of Genetics Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Epidemiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jean-François Zagury
- Laboratoire Génomique, Bioinformatique, et Applications, EA4627, Chaire de Bioinformatique, Conservatoire National des Arts et Métiers, Paris, France
- ANRS Genomic Group (French Agency for Research on AIDS and Hepatitis), Paris, France
| | - Jacques Fellay
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
50
|
Corchado S, Márquez M, Montes de Oca M, Romero-Cores P, Fernández-Gutiérrez C, Girón-González JA. Influence of Genetic Polymorphisms of Tumor Necrosis Factor Alpha and Interleukin 10 Genes on the Risk of Liver Cirrhosis in HIV-HCV Coinfected Patients. PLoS One 2013; 8:e66619. [PMID: 23840511 PMCID: PMC3694087 DOI: 10.1371/journal.pone.0066619] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 05/09/2013] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Analysis of the contribution of genetic (single nucleotide polymorphisms (SNP) at position -238 and -308 of the tumor necrosis factor alpha (TNF-α) and -592 of the interleukin-10 (IL-10) promotor genes) and of classical factors (age, alcohol, immunodepression, antirretroviral therapy) on the risk of liver cirrhosis in human immunodeficiency (HIV)-hepatitis C (HCV) virus coinfected patients. PATIENTS AND METHODS Ninety one HIV-HCV coinfected patients (50 of them with chronic hepatitis and 41 with liver cirrhosis) and 55 healthy controls were studied. Demographic, risk factors for the HIV-HCV infection, HIV-related (CD4+ T cell count, antiretroviral therapy, HIV viral load) and HCV-related (serum ALT concentration, HCV viral load, HCV genotype) characteristics and polymorphisms at position -238 and -308 of the tumor necrosis factor alfa (TNF- α) and -592 of the interleukin-10 (IL-10) promotor genes were studied. RESULTS Evolution time of the infection was 21 years in both patients' groups (chronic hepatitis and liver cirrhosis). The group of patients with liver cirrhosis shows a lower CD4+ T cell count at the inclusion in the study (but not at diagnosis of HIV infection), a higher percentage of individuals with previous alcohol abuse, and a higher proportion of patients with the genotype GG at position -238 of the TNF-α promotor gene; polymorphism at -592 of the IL-10 promotor gene approaches to statistical significance. Serum concentrations of profibrogenic transforming growth factor beta1 were significantly higher in healthy controls with genotype GG at -238 TNF-α promotor gene. The linear regression analysis demonstrates that the genotype GG at -238 TNF-α promotor gene was the independent factor associated to liver cirrhosis. CONCLUSION It is stressed the importance of immunogenetic factors (TNF-α polymorphism at -238 position), above other factors previously accepted (age, gender, alcohol, immunodepression), on the evolution to liver cirrhosis among HIV-infected patients with established chronic HCV infections.
Collapse
Affiliation(s)
- Sara Corchado
- Unidad de Enfermedades Infecciosas, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Mercedes Márquez
- Unidad de Enfermedades Infecciosas, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | | | - Paula Romero-Cores
- Unidad de Enfermedades Infecciosas, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | | | | |
Collapse
|